The p40phox protein, a regulatory component of the phagocyte NADPH oxidase, is preferentially expressed in cells of myeloid lineage. We investigated transcriptional regulation of thep40phox gene in HL-60 myeloid cells. Deletion analysis of approximately 6 kb of the 5′-flanking sequence of the gene demonstrated that the proximal 106 base pair of the promoter exhibited maximum reporter activity. This region contains 3 potential binding sites for PU.1, a myeloid-restricted member of theets family of transcription factors. Mutation or deletion of each PU.1 site decreased promoter activity, and the level of activity mediated by each site correlated with its binding avidity for PU.1, as determined by gel shift competition assays. Mutation of all 3 sites abolished promoter activity in myeloid cells. PU.1-dependent expression was also observed in the Raji B-cell line, whereas the moderate level of promoter reporter activity in the nonmyeloid HeLa cell line was independent of PU.1. Chromatin immunoprecipitation assay demonstrated occupation of the PU.1 sites by PU.1 in vivo in HL-60 cells. Cotransfection of the pGL3-p40-106 reporter construct with a dominant-negative PU.1 mutant dramatically reduced promoter activity, whereas the overexpression of PU.1 increased promoter activity. Promoter activity and transcript levels ofp40phox increased in HL-60 cells during dimethyl sulfoxide–induced differentiation toward the granulocyte phenotype, and this was associated with increased cellular levels of PU.1 protein. Our findings demonstrate that PU.1 binding at multiple sites is required for p40phox gene transcription in myeloid cells and that granulocytic differentiation is associated with the coordinated up-regulation of PU.1 andp40phox expression.

NADPH oxidase is the major inducible source of superoxide and superoxide-derived reactive oxygen species in phagocytes and is a key enzyme in host defense against microbial infection.1-3 Genetic deficiency of this enzyme results in the inherited disorder chronic granulomatous disease (CGD), which is characterized by impaired phagocyte microbicidal activity and a clinical syndrome of recurrent life-threatening infections.1-3 The reactive oxygen species generated by this enzyme are also toxic to host cells and may account for much of the tissue injury that occurs during inflammation.4-6Therefore, the expression and activity of the phagocyte NADPH oxidase components must be tightly regulated.

The fully assembled active NADPH oxidase complex is composed of membrane-bound and cytosolic components.7,8 The membrane-associated core component of the oxidase is a heterodimeric b-type cytochrome comprised of light (p22phox) and heavy (gp91phox) chain polypeptides. Cytosolic components include p67phox, p47phox, p40phox, and the small GTPase protein, Rac2. X-linked CGD, the most prevalent form of the disorder, is a result of mutations in the gp91phox gene, whereas autosomal recessive CGD is associated with mutations of thep47phox, p67phox, orp22phox genes.1-3 

Numerous studies have demonstrated essential roles for gp91phox, p22phox, p67phox, p47phox, and Rac2 in phagocyte NADPH oxidase function.9-20Reconstitution of the oxidase by transfection into cell lines deficient in the oxidase proteins or recombination of the components in vitro shows that efficient superoxide generation by the NADPH oxidase complex is absolutely dependent on these components. The role of p40phox, on the other hand, is less clearly defined because it does not appear to be absolutely necessary for the core enzymatic function, namely the univalent reduction of oxygen. Nevertheless, several lines of evidence suggest that p40phox is directly involved in the normal physiologic function of the oxidase. Thus, p40phox forms a trimolecular complex with p47phox and p67phoxin the cytosol of resting phagocytes and on activation translocates to the membrane with these components.21-23 Moreover, p40phox can bind p67phox and p47phox, and the strong binding of p40phox to p67phox can be disrupted by the activated guanosine triphosphate-bound form of Rac, a key intermediate in oxidase activation.23-31 The SH3 domain of p40phox competes for the C-terminal, proline-rich domain in p47phox, which also interacts with the C-terminal SH3 domain of p67phox and thereby down-regulates NADPH oxidase activity.24,25,29,30 In contrast, other studies suggest that p40phox actually participates in the activation of NADPH oxidase by increasing the affinity of p47phox for the flavocytochrome.32More recently, it has been reported that the 57-kd actin-binding protein coronin associates with the p40phox–p67phox–p47phoxcytosolic complex through the C-terminal domain of p40phox and that it colocalizes with the phox proteins in the periphery of the phagocytic vacuole during phagocytosis and activation of the oxidase.33Finally, recent studies have shown that p40phox is hyperphosphorylated during NADPH oxidase activation and dephosphorylated during enzyme inactivation.34 35 In summary, these studies suggest that p40phox acts as a regulator of oxidase activity and as an adapter that links oxidase to the cytoskeletal elements that participate in cell activation.

Tissue distribution studies have demonstrated that, similar to the gp91phox, p47phox, and p67phox components of NADPH oxidase, p40phox is preferentially expressed in cells of myeloid lineage.36,37 Transcription of many myeloid-specific genes is regulated by PU.1, a member of theets family of transcription factors.38Gene-targeting studies have demonstrated that PU.1 is essential for the normal embryonic development of hematopoietic cells of myeloid, B-cell, and T-cell lineages.39,40 We have previously characterized the promoter of the p47phox gene and demonstrated that PU.1 is essential for p47phoxtranscription in myeloid cells.41 PU.1 appears to regulate transcription by binding to a single cis element located in the proximal region of the p47phox gene. In this report, we describe our studies on the transcriptional regulation of the p40phox gene in the HL-60 myeloid cell line. Our data show that p40phoxtranscription is regulated by 3 PU.1 binding sites located within the proximal region of the p40phox promoter and that the PU.1 transcription factor binds to each site, in vitro and in vivo. The central role of PU.1 in the regulation ofp40phox expression supports previous observations that this phox protein is mainly restricted to cells of myeloid lineage and is of particular importance in phagocyte function.

Materials

RPMI 1640 was obtained from Life Technologies (Gaithersburg, MD). Restriction enzymes, T4 polynucleotide kinase, RNasin, and pGL3-Basic luciferase reporter vector and the dual luciferase assay kit were from Promega (Madison, WI). γ-[32P]Adenosine triphosphate (ATP), 6000 Ci/mmol (370 MBq/mol), was obtained from DuPont-NEN (Boston, MA). The TOPO-TA Cloning Kit, with the pCRII vector for cloning products of the polymerase chain reaction (PCR), was obtained from Invitrogen (San Diego, CA). Oligonucleotides were synthesized at the Advanced DNA Technology Unit (University of Texas Health Science Center at San Antonio). The Sequenase DNA sequencing kit was obtained from United States Biochemical (Cleveland, OH). PU.1 antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA).

Human p40phoxgenomic cloning and sequencing

The p40phox 5′-flanking region was cloned using the PromoterFinder Kit (Clontech, Palo Alto, CA) according to the manufacturer's protocol. PCR was performed using the human genomic libraries provided as templates to amplify the desired sequences. The forward primer was complementary to the adaptor ligated to the genomic DNA fragments contained in each library. The reverse primer (5′-CCAGGGAGCAGGTGGAGAGTCTCGC-3′, Figure 2A) was complementary to base pair (bp) +142 to +117 of thep40phox gene.22 36Amplified products were analyzed on a 1.2% agarose gel and then subjected to a second round of PCR with nested primers. The nested reverse primer (5′-AGGCTGAGTTCACCTCTCACTTCC-3′) was complementary to bp +105 to +81 of the gene. Final PCR products were cloned directly into the pCRII vector, and their identities were confirmed by sequencing. Nucleotide sequencing was carried out in both directions using the dideoxy termination procedure and sequence-specific oligonucleotide primers.

Luciferase vector construction

Reporter vectors were constructed in the pGL3-Basic luciferase vector. Promoter regions were amplified using human genomic DNA as template. The reverse primer was similar to the nested reverse primer used above, but a site for cleavage by XhoI was added (5′-CACTCGAGAGGCTGAGTTCACCTCTC-3′). The forward primers corresponded to the upstream sequences of desired promoter regions, with a KpnI cleavage site added at the 5′ end. PCR products were digested with KpnI and XhoI and were cloned into the pGL3-Basic reporter vector. Inserts of the constructs generated all extend downstream to +104 relative to the transcription start site of the p40phoxgene.36 Mutations of the PU.1 binding sites were generated by site-directed mutagenesis (QuikChange kit; Stratagene, La Jolla, CA) using the primers illustrated in Figure 2B. Constructs were confirmed by restriction mapping and sequencing.

Cell culture and transient transfections

As previously described,41 the human myeloid cell lines HL-60, THP-1, U937, and PLB985 and the human B-cell line Raji were maintained in RPMI 1640 supplemented with 10% fetal bovine serum, 10 mM HEPES, penicillin, and streptomycin. Transfection was carried out by electroporation as we have described.41 Briefly, approximately 107 cells were resuspended in 0.5 mL complete medium containing 20 μg luciferase reporter constructs and 0.2 μg of a Renilla luciferase vector (pRL-null; Promega) as a transfection efficiency control. In Figure 1, transfection was also carried out with equimolar amounts (7 pmol) of each construct, plus an unrelated plasmid, pUC19, to provide an equal amount of total DNA. For cotransfection assays, 10 μg each reporter construct and PU.1 expression plasmid (a generous gift from Dr M. Klemsz, Indiana University, Indianapolis) were used. Electroporation was carried out at 960 μF and 250 V for HL-60 cells or 320 V for Raji cells. At 48 hours the cells were washed 3 times in phosphate-buffered saline (pH 7.4), lysed in 100 μL 1× reporter lysis buffer (Promega), and centrifuged at 400g for 5 minutes at ambient temperature, and 20-μL aliquots of the supernatants were tested for reporter gene activity using the dual luciferase assay system (Promega) and a Turner Designs TD–20/20 luminometer. The human cervical carcinoma epithelial cell line HeLa was grown in Dulbecco modified Eagle medium supplemented with 10% fetal bovine serum and transfected by Lipofectamine Plus reagent (Gibco) according to the manufacturer's protocol.

Fig. 1.

Identification of the proximal promoter region of thep40phox gene.

HL-60 cells in log phase of growth were transfected with the indicated constructs and assayed for luciferase activity after 48 hours. Allp40phox constructs extended from the indicated position in the 5′-flanking sequence of the gene to nucleotide +104 of the 5′-UTR relative to the reported transcriptional start site.36 Luciferase activity is reported as the ratio of the test construct to the promoterless vector pGL3-Basic. Values were corrected for transfection efficiency by cotransfection with the renilla expression plasmid and were normalized to equal molar content of DNA. Data shown are means (± SE) of 5 independent experiments.

Fig. 1.

Identification of the proximal promoter region of thep40phox gene.

HL-60 cells in log phase of growth were transfected with the indicated constructs and assayed for luciferase activity after 48 hours. Allp40phox constructs extended from the indicated position in the 5′-flanking sequence of the gene to nucleotide +104 of the 5′-UTR relative to the reported transcriptional start site.36 Luciferase activity is reported as the ratio of the test construct to the promoterless vector pGL3-Basic. Values were corrected for transfection efficiency by cotransfection with the renilla expression plasmid and were normalized to equal molar content of DNA. Data shown are means (± SE) of 5 independent experiments.

Close modal

In vitro synthesis

Mouse PU.1 cDNA42 was subcloned into pBluescript SK and was translated in vitro using T3 RNA polymerase and the TnT-coupled reticulocyte lysate system (Promega). Synthesized [35S]-methionine–labeled PU.1 was analyzed by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and fluorography. A predominant band measuring approximately 38 kd was observed, consistent with the molecular mass previously reported.43 The control un-programmed sample (ie, no cDNA) produced no corresponding band.

Nuclear extracts

Cells were disrupted by nitrogen cavitation using a technique originally described for neutrophils,44 and nuclear extracts were prepared as we have previously reported in detail.41 Extracts were collected and stored in aliquots at −70°C. The protein concentration was determined using the Bradford reagent (Bio-Rad, Hercules, CA).

Electrophoretic mobility shift assay

Complementary DNA oligonucleotides were annealed by heating in 1× NET at 95°C for 5 minutes and cooling at ambient temperature. Probes were then labeled with [γ-32P]ATP and T4 polynucleotide kinase. For gel shift assays, nuclear extract (6 μg) was incubated for 20 minutes at ambient temperature with 5 × 104 cpm of the labeled DNA probe in 20 μL binding buffer containing 10 mM Tris-HCl (pH 7.6), 50 mM NaCl, 1 mM EDTA, 1 mM dithiothreitol, 5% glycerol, 1 μg/μL bovine serum albumin, and 2 μg poly-d(I-C). For supershift assays, 2 μL specific antibody was added, and the reaction was continued for 15 minutes. Samples were loaded on 5% nondenaturing polyacrylamide gels, and electrophoresis was carried out at 200 V in 25 mM Tris (pH 8.5) with 190 mM glycerol and 1 mM EDTA. Competition assays were carried out in the same manner, except that the above reaction mixture was preincubated with competitor DNA for 10 minutes at 4°C before the labeled probe was added. Signals were quantitated with a PhosphorImager and ImageQuant software (Molecular Dynamics). Relative binding avidity of various DNA probes for PU.1 were determined by comparisons of band intensities observed in the presence of varied amounts of probes or a series of dilutions of competing oligonucleotides.

Chromatin immunoprecipitation analysis

Chromatin immunoprecipitation (ChIP) analysis was carried out as described by Boyd et al.45 Briefly, formaldehyde was added directly to the cell culture to a final concentration of 1% (wt/vol), and the cells were incubated at 23°C for 10 minutes. Glycine was added to stop the reaction, and the cells were collected, washed, and allowed to swell on ice for 10 minutes in 5 mM PIPES (pH 8.0) containing 85 mM KCl, 0.5% NP-40, 0.5 mM phenylmethylsulfonyl fluoride, and 100 ng/mL leupeptin and aprotinin. Pelleted nuclei were collected and incubated on ice for 10 minutes in 50 mM Tris HCl (pH 8.1), 1% SDS, 10 mM EDTA, and the protease inhibitors, sonicated on ice to break the chromatin DNA, and precleared with Staph A cells (protein A–positive Staphylococcus aureus cells; Roche Molecular Biochemicals, Indianapolis, IN). Precleared chromatin from 2 × 107 cells was incubated with 1.5 μg affinity-purified rabbit polyclonal antibody to PU.1 (anti-PU.1 sc-352X; Santa Cruz Biotechnology) or without antibody, and it was rotated at 4°C for 12 to 16 hours. Immunoprecipitation was carried out with Staph A cells. The supernatant from the reaction lacking primary antibody was saved as total input of chromatin and was processed in the same way as the eluted immunoprecipitates, starting with the cross-link reversal step. Cross-linking was reversed by incubation at 65°C for 5 hours in the presence of 300 mM NaCl and 0.01% RNase A. Samples were then precipitated, resusupended, and treated with proteinase K, followed by extraction with phenol-chloroform-isoamyl alcohol and precipitation with sodium acetate and ethanol plus tRNA and glycogen as carriers. Pellets were collected by microcentrifugation, resuspended in 30 μL H2O, and analyzed by PCR. Total input samples were resuspended in 100 μL H2O and then diluted 1:100 before PCR. PCR reactions were carried out in a total volume of 25 μL containing 2-3 μL immunoprecipitate or diluted total input as the template and using recombinant Taq DNA polymerase (Life Technologies). After 35 cycles of amplification, PCR products were separated on 1.5% agarose containing ethidium bromide. PCR primers (p40-PU.1 forward, 5′-GCGCCAAGGACTGACATC-3′ and reverse, 5′-GAGCAGGTGGTGCGTCTC-3′) were designed to amplify a 299-bp region of the p40phox promoter that contains the 3 PU.1 sites.

Northern blot analysis

Total cellular RNA was isolated using the Ultraspec reagent (Biotecx Laboratories, Houston, TX) and dissolved in nuclease-free water. Samples of RNA (10 μg) were separated by electrophoresis in 1% agarose under denaturing conditions (formaldehyde), transferred to Nytran Plus membrane (Schleicher & Schuell, Keene, NH), UV cross-linked, and probed with p40phoxcDNA labeled using the High Prime DNA labeling kit, (Roche Molecular Biochemicals) and [α-32P]-dCTP (DuPont-NEN). Following autoradiography at −70°C, the membrane was stripped and probed with32P-labeled cDNA for human acidic ribosomal phosphoprotein 36B4 as a loading control.

The 5′-flanking region of the humanp40phoxgene exhibits promoter activity in myeloid cells

Previous studies have shown that humanp40phox mRNA is expressed almost exclusively in hematopoietic cells that exhibit NADPH oxidase activity.36,37 To identify the sites of promoter activity in the p40phox gene, regions of the 5′-flanking sequence were prepared by PCR, cloned into the luciferase reporter vector pGL3-Basic, and assayed by transient transfection and expression in the promyelocytic HL-60 cell line. The highest level of activity (approximately 37-fold increase over pGL3-Basic vector control) was observed with the construct pGL3-p40-106 (Figure1), which contained the first 106 nucleotides upstream of the p40phoxtranscription start site.36 Constructs extending further upstream exhibited progressively lower luciferase activity, suggesting that the minimal p40phox promoter lies within the first 106 nucleotides of the 5′-flanking sequence of thep40phox gene and that negative regulatory elements may be present in upstream regions. Other myeloid cell lines tested (THP-1, U937, and PLB985) showed similar results (data not shown).

Three PU.1 consensus sites in the proximalp40phoxpromoter bind PU.1

Inspection of the p40phox promoter for potential regulatory elements identified several core-binding sequences (GAGGAA) for the PU.1 transcription factor. Applying criteria for optimal flanking nucleotides that we identified previously,46 3 of these PU.1 sites (PU.1a, −95 to −100; PU.1b, −65 to −60; and PU.1c, +80 to +85; Figure2A) were considered to have a high probability of binding PU.1. To determine whether these sites bound PU.1, electrophoretic mobility shift assay (EMSA) was carried out using32P-labeled double-stranded oligonucleotides corresponding to these regions (Figure 2B) and nuclear extracts of HL-60 cells. Several DNA–protein complexes were formed between the PU.1 probes and HL-60 nuclear extracts (Figure 2C). Effective competition with excess unlabeled wild-type, but not mutated, DNA probes demonstrated that these bands were specific. The addition of specific antibody to PU.1 resulted in a dramatic decrease in band intensity and in the appearance of new supershifted bands, providing clear evidence that the complexes contained PU.1 protein. Comparison with previous studies suggested that the major bands of slower mobility contained intact PU.1 protein, whereas the faster-migrating bands were probably formed by PU.1 degradation products.46 

Fig. 2.

Characterization of 3 PU.1 binding sites in the p40phox promoter.

(A) Sequence of the proximal promoter region of thep40phox gene. 3 PU.1 sites are underlined and labeled PU.1a, PU.1b, and PU.1c. The arrow indicates the reported transcription start site,36 and the translation initiation codon is double-underlined. Nested reverse primers used in cloning are in bold font. (B) Sequences of wild-type and mutatedp40phox PU.1 oligonucleotides used in the EMSA studies. Core PU.1 binding domains are underlined, and mutated nucleotides are shown in bold. (C) EMSA of HL-60 nuclear extracts with32P-labeled p40phox PU.1 DNA probes. HL-60 nuclear extract (5 μg) was incubated with the labeled probes alone (lanes 1, 5, and 9) or together with either antibody to PU.1 (lanes 2, 6, and 10) or a 200-fold molar excess of the homologous wild-type (lanes 3, 7, and 11) or mutated (lanes 4, 8, and 12) oligonucleotides (Figure 2B). The specific PU.1-DNA complex (PU.1) and the supershifted complex (SS) are indicated by arrows.

Fig. 2.

Characterization of 3 PU.1 binding sites in the p40phox promoter.

(A) Sequence of the proximal promoter region of thep40phox gene. 3 PU.1 sites are underlined and labeled PU.1a, PU.1b, and PU.1c. The arrow indicates the reported transcription start site,36 and the translation initiation codon is double-underlined. Nested reverse primers used in cloning are in bold font. (B) Sequences of wild-type and mutatedp40phox PU.1 oligonucleotides used in the EMSA studies. Core PU.1 binding domains are underlined, and mutated nucleotides are shown in bold. (C) EMSA of HL-60 nuclear extracts with32P-labeled p40phox PU.1 DNA probes. HL-60 nuclear extract (5 μg) was incubated with the labeled probes alone (lanes 1, 5, and 9) or together with either antibody to PU.1 (lanes 2, 6, and 10) or a 200-fold molar excess of the homologous wild-type (lanes 3, 7, and 11) or mutated (lanes 4, 8, and 12) oligonucleotides (Figure 2B). The specific PU.1-DNA complex (PU.1) and the supershifted complex (SS) are indicated by arrows.

Close modal

Intact PU.1 binding sites are required for promoter activity in HL-60 myeloid cells and Raji B cells, but not in nonmyeloid HeLa cells

To investigate the role of these 3p40phox PU.1 binding sites on promoter activity, a series of mutation and deletion promoter–luciferase constructs were prepared in which the 3 PU.1 sites were deleted or mutated individually or in combination. These constructs were tested as before by transient expression in HL-60 cells. Mutation of PU.1a, PU.1b, or PU.1c in the −1599 bpp40phox promoter construct reduced activity by 82%, 63%, and 18%, respectively (Figure3A). Similar results were observed with the deletion series (Figure 3B). The p40phoxpromoter activity in the −1599 bp construct was nearly abolished in the HL-60 cells when all 3 binding sites were mutated, indicating the essential requirement for PU.1 in this cell type (Figure 3B). Because PU.1 and p40phox are also expressed in B lymphocytes, we investigated the roles of the PU.1 sites on p40phox promoter activity in this cell type using the Raji human B-cell line. The PU.1 sites in thep40phox promoter bound to PU.1 protein in Raji nuclear extracts (Figure 4A). Furthermore, the promoter was active in these cells, though at levels lower than those observed in the HL-60 cells (Figure 4B). More important, however, the promoter function was dependent on the presence of intact PU.1a and PU.1b sites (Figure 4B; PU.1c not tested). To investigate further the requirement for the PU.1 sites inp40phox promoter activity, the mutated constructs were also analyzed in the nonmyeloid HeLa cell line, which does not express PU.1. Although the activity of the wild type −1599 bpp40phox promoter construct was high in HeLa (Figure 4C), this activity was not affected by mutation of the PU.1 sites. The relatively high basal activity seen in HeLa cells with these constructs was surprising and might have been mediated by transcription factors present in these cells that bind the −1599p40phox promoter construct at sites distinct from the PU.1 elements. These sites might normally be repressed in nonmyeloid tissues in vivo through elements in the gene not included in the −1599 p40phoxconstruct. Such factors would be absent, functionally inactive, or PU.1-dependent in HL-60 and Raji cells because in both cases, mutation of the PU.1 sites was sufficient to eliminate most promoter activity.

Fig. 3.

Effect of 3 PU.1 sites onp40phox promoter function in HL-60 cells.

(A) Mutational analysis of the −1599 to +104 region of thep40phox promoter. The 3 PU.1 sites (open boxes) present in the pGL3-p40-1599 construct were mutated (hatched boxes) singly or in combination, and the resultant constructs were assayed for reporter gene activity in HL-60 cells as in Figure 1. The arrow indicates the reported transcription start site. (B) Deletion analysis of the contribution of each PU.1 site to the overall transactivation activity of the proximal promoter (−106 to +104 bp) of the p40phox gene. Deletion constructs were prepared and assayed for reporter gene activity in HL-60 cells as in Figure 1. Data shown in both panels are means (± SE) of 8 independent experiments. Analysis of variance showed that differences in luciferase activity among the constructs were significant (P < .01), and t test demonstrated that the luciferase activities of the mutation and deletion constructs were significantly lower (P < .01 except for the PU.1c mutation construct, where P = .044) than those from wild-type counterparts.

Fig. 3.

Effect of 3 PU.1 sites onp40phox promoter function in HL-60 cells.

(A) Mutational analysis of the −1599 to +104 region of thep40phox promoter. The 3 PU.1 sites (open boxes) present in the pGL3-p40-1599 construct were mutated (hatched boxes) singly or in combination, and the resultant constructs were assayed for reporter gene activity in HL-60 cells as in Figure 1. The arrow indicates the reported transcription start site. (B) Deletion analysis of the contribution of each PU.1 site to the overall transactivation activity of the proximal promoter (−106 to +104 bp) of the p40phox gene. Deletion constructs were prepared and assayed for reporter gene activity in HL-60 cells as in Figure 1. Data shown in both panels are means (± SE) of 8 independent experiments. Analysis of variance showed that differences in luciferase activity among the constructs were significant (P < .01), and t test demonstrated that the luciferase activities of the mutation and deletion constructs were significantly lower (P < .01 except for the PU.1c mutation construct, where P = .044) than those from wild-type counterparts.

Close modal
Fig. 4.

PU.1 is present in nuclear extracts from HL-60 and Raji cells, binds to the p40phoxpromoter, and is required for reporter activity in Raji, but not HeLa cells.

(A) EMSA of nuclear extracts with 32P-labeledp40phox PU.1a DNA probe. HeLa (lanes 1-3), HL-60 (lanes 4-6) or Raji (lanes 7-12) nuclear extracts (5 μg) were incubated with the labeled probes alone (lanes 2, 5, and 8) or together with either antibody to PU.1 (lane 10) or a 200-fold molar excess of unlabeled PU.1a (lanes 3, 6, and 9), PU.1b (lane 11), or PU.1c (lane 12) oligonucleotide (see Figure 2B). The specific PU.1-DNA complex (PU.1) and the supershifted complex (SS) are indicated by arrows. Mutational analysis of the −1599 to +104 region of thep40phox promoter in Raji (B) or HeLa cells (C). The PU.1 sites (open boxes) present in the pGL3-p40-1599 construct were mutated (hatched boxes), and the resultant constructs were assayed for reporter gene activity as in Figure 1. Arrow indicates the reported transcription start site. Data shown are means (± SE) of 3 or more independent experiments.

Fig. 4.

PU.1 is present in nuclear extracts from HL-60 and Raji cells, binds to the p40phoxpromoter, and is required for reporter activity in Raji, but not HeLa cells.

(A) EMSA of nuclear extracts with 32P-labeledp40phox PU.1a DNA probe. HeLa (lanes 1-3), HL-60 (lanes 4-6) or Raji (lanes 7-12) nuclear extracts (5 μg) were incubated with the labeled probes alone (lanes 2, 5, and 8) or together with either antibody to PU.1 (lane 10) or a 200-fold molar excess of unlabeled PU.1a (lanes 3, 6, and 9), PU.1b (lane 11), or PU.1c (lane 12) oligonucleotide (see Figure 2B). The specific PU.1-DNA complex (PU.1) and the supershifted complex (SS) are indicated by arrows. Mutational analysis of the −1599 to +104 region of thep40phox promoter in Raji (B) or HeLa cells (C). The PU.1 sites (open boxes) present in the pGL3-p40-1599 construct were mutated (hatched boxes), and the resultant constructs were assayed for reporter gene activity as in Figure 1. Arrow indicates the reported transcription start site. Data shown are means (± SE) of 3 or more independent experiments.

Close modal

Promoter activity of the PU.1 sites correlates with PU.1 binding avidity

Our previous studies indicated a correlation between the transactivation activity of PU.1 sites from a number of promoters and their affinity for the PU.1 protein.46 Thus, we hypothesized that the different levels of transactivation activity of the PU.1 sites in the p40phox promoter might be attributed in part to their relative PU.1 binding avidity. To investigate this possibility, the binding affinities of the 3 sites for PU.1 present in HL-60 nuclear extracts or in vitro synthesized PU.1 were estimated by 2 methods. First, increasing amounts of each labeledp40phox PU.1 oligonucleotide probe were incubated with a fixed amount of PU.1 protein (Figure5A). Second, the ability of unlabeledp40phox PU.1 oligonucleotide probes to compete for binding with the well-characterizedp47phox PU.1 site41 46 was determined (Figure 5B). Semiquantitative analysis of these binding studies suggested that the binding avidity of the PU.1a site is 2- to 4-fold greater than that of the PU.1b site (compare lane 2 with lanes 8 and 9 in Figure 5A and lane 2 with lane 6 in Figure 5B). The binding avidity of PU.1b is, in turn, 3- to 8-fold greater than that of the PU.1c site (compare lane 7 with lane 15 in Figure 5A and lane 5 with lane 9 in Figure 5B). Combining these EMSA results with the data obtained by deletion and mutation transfection studies indicated a direct relationship between the avidity with which these sites bind PU.1 and their capacity to dictate reporter gene transcription (PU.1a is greater than PU.1b is greater than PU.1c). However, the decrease in promoter activity on deletion of the PU.1c site was 20% when the other 2 PU.1 binding sites were intact, but it was 36% when the PU.1a site was deleted and 45% when the PU.1a and PU.1b sites were deleted (data not shown), suggesting that compensation may take place when one or another of the PU.1 sites is inactivated.

Fig. 5.

The p40phox PU.1 sites bind PU.1 with different avidity.

(A) EMSA of the p40phox PU.1 sites with varied amounts of the DNA probes. Increasing amounts of the32P-labeled PU.1 DNA probes having comparable specific activities of labeling were incubated with in vitro–synthesized PU.1 protein and then analyzed by PAGE. The major PU.1-DNA complex is indicated by the arrow. (B) Competition between oligonucleotides corresponding to the p40phox PU.1 binding sites and a 32P-labeled PU.1 DNA probe (5′-CAAAAGCGACTTCCTCTTTCCAGTGC-3′) from thep47phox promoter for binding to PU.1 protein in HL-60 nuclear extracts. Fixed amounts of HL-60 nuclear extract and 32P-labeledp47phox PU.1 probe were incubated in the presence of the indicated amounts of the unlabeledp40phoxoligonucleotides. Specificity of the PU.1-DNA complex indicated by the arrow was confirmed by supershift assay with antibodies to human PU.1 (data not shown).

Fig. 5.

The p40phox PU.1 sites bind PU.1 with different avidity.

(A) EMSA of the p40phox PU.1 sites with varied amounts of the DNA probes. Increasing amounts of the32P-labeled PU.1 DNA probes having comparable specific activities of labeling were incubated with in vitro–synthesized PU.1 protein and then analyzed by PAGE. The major PU.1-DNA complex is indicated by the arrow. (B) Competition between oligonucleotides corresponding to the p40phox PU.1 binding sites and a 32P-labeled PU.1 DNA probe (5′-CAAAAGCGACTTCCTCTTTCCAGTGC-3′) from thep47phox promoter for binding to PU.1 protein in HL-60 nuclear extracts. Fixed amounts of HL-60 nuclear extract and 32P-labeledp47phox PU.1 probe were incubated in the presence of the indicated amounts of the unlabeledp40phoxoligonucleotides. Specificity of the PU.1-DNA complex indicated by the arrow was confirmed by supershift assay with antibodies to human PU.1 (data not shown).

Close modal

PU.1 binds to the p40phoxpromoter in vivo

To determine whether PU.1 protein binds thep40phox promoter at the identified PU.1 sites in vivo, we performed ChIP analysis on HL-60 cells using a specific polyclonal antibody to PU.1. Following immunoprecipitation and reversal of cross-linking, the DNA was purified and used as a template for PCR amplification using primers that encompass the region of thep40phox promoter containing the 3 PU.1 binding sites. As a negative control, we carried out immunoprecipitation with antibody to c-Jun, consensus binding sites for which are absent from this region of thep40phox promoter. Thep40phox primers produced an amplicon of predicted size from the total input DNA and the anti–PU.1-precipitated DNA, but not from anti–c-Jun–precipitated DNA (Figure6). Furthermore, PCR primers for an unrelated gene, CCR5, generated products only from the total input DNA, confirming the specificity of the assay. The CCR5primers were designed to amplify a region of the CCR5gene that does not contain PU.1 or c-Jun binding sites. These data indicate that endogenous PU.1 protein in HL-60 cells binds thep40phox promoter in the region containing the PU.1 sites and thus may promote transcription of the gene in vivo.

Fig. 6.

ChIP analysis of thep40phox promoter PU.1-binding sites.

Cross-linked HL-60 chromatin was immunoprecipitated with antibodies to PU.1 or c-Jun, or in the absence of antibody (Input). Cross-linking was reversed, and the DNA was purified and analyzed by PCR with primers designed to amplify a p40phox DNA fragment containing these PU.1-binding sites or a segment of theCCR5 promoter that does not contain PU.1-binding sites.

Fig. 6.

ChIP analysis of thep40phox promoter PU.1-binding sites.

Cross-linked HL-60 chromatin was immunoprecipitated with antibodies to PU.1 or c-Jun, or in the absence of antibody (Input). Cross-linking was reversed, and the DNA was purified and analyzed by PCR with primers designed to amplify a p40phox DNA fragment containing these PU.1-binding sites or a segment of theCCR5 promoter that does not contain PU.1-binding sites.

Close modal

Overexpression of PU.1 increases p40phoxpromoter activity, whereas a dominant-negative mutant of PU.1 decreases activity

To provide further evidence that PU.1 is critical forp40phox promoter activity, thep40phox luciferase construct pGL3-p40-106 or pGL-3-Basic control were transfected into HL-60 cells with or without cotransfection of either the PU.1 expression plasmid pJ6-mPU.1 or the dominant-negative mutant plasmid pJ6-NN, in which the transactivation domain is deleted. Overexpression of PU.1 increased the activity of the p40phox promoter by approximately 2.5-fold (Figure 7), whereas expression of the dominant-negative mutant PU.1 decreased activity by approximately 50%, presumably because of competition with endogenous PU.1. That the transactivation was dependent on the binding of PU.1 to the putative PU.1 sites in the proximal portion of thep40phox promoter was confirmed in control experiments using the pGL3-p40-106mt construct, in which all 3 PU.1 sites were mutated. No effect of wild-type or mutant PU.1 was observed with this mutant reporter construct (Figure 7).

Fig. 7.

Effect of exogenous PU.1 onp40phox promoter function.

HL-60 cells were transfected with the wild-type reporter vector pGL3-p40-106 or the analogous vector in which all 3 PU.1 sites were mutated (pGL3-p40-106-mtPU.1) and were cotransfected with either a wild-type PU.1 expression plasmid (pJ6-mPU.1), a dominant-negative PU.1 mutant plasmid (pJ6-NN), or the empty expression vector (pJ6). Reporter gene activity was assayed as before, and results were expressed as the mean (± SE) of 3 independent experiments.

Fig. 7.

Effect of exogenous PU.1 onp40phox promoter function.

HL-60 cells were transfected with the wild-type reporter vector pGL3-p40-106 or the analogous vector in which all 3 PU.1 sites were mutated (pGL3-p40-106-mtPU.1) and were cotransfected with either a wild-type PU.1 expression plasmid (pJ6-mPU.1), a dominant-negative PU.1 mutant plasmid (pJ6-NN), or the empty expression vector (pJ6). Reporter gene activity was assayed as before, and results were expressed as the mean (± SE) of 3 independent experiments.

Close modal

Induction of granulocyte differentiation in HL-60 cells increases PU.1 protein, p40phoxmRNA expression, and p40phoxpromoter activity

HL-60 cells can be induced to differentiate toward a granulocytic phenotype by treatment with dimethyl sulfoxide (DMSO).47Components of the phagocyte NADPH oxidase system are induced by DMSO, and enzymatic activity can be detected within 1 to 2 days of treatment48-50 Althoughp40phox is not an essential component of the functional NADPH oxidase, it is expressed predominantly in phagocytic cells, complexed with the essential oxidase components p47phox and p67phox, and, in all probability, has a role in enzyme activation and regulation.29 32 To investigate the influence of granulocytic differentiation on p40phoxgene expression, we first examined the induction ofp40phox mRNA during DMSO treatment. Total RNA was isolated from DMSO-treated HL-60 cells at daily intervals, and Northern blot analysis was carried out using a cDNA probe specific for human p40phox. Levels of p40phox mRNA increased approximately 8-fold within 1 day of treatment (Figure8A). Subsequently, transcript levels decreased but were maintained at approximately 4-fold over controls for 2 to 3 days. To determine whether increased gene transcription contributed to this observed increase inp40phox gene expression, HL-60 cells were first transfected with the pGL-3–p40-106 reporter gene construct, then treated with DMSO for 38 hours, and luciferase reporter activity was measured. DMSO increased reporter gene activity by 3- to 4-fold over the untreated control (Figure 8B). However, when we transfected the construct in which all 3 PU.1 sites were mutated, only a small increase in luciferase activity was seen following DMSO treatment. This suggests that the increase in transcriptional activity induced by DMSO in the wild-type promoter was mediated through increased PU.1 activity. These data were supported by EMSA and immunoblot analysis because nuclear extracts from HL-60 cells taken 38 hours after DMSO treatment showed increased PU.1 binding activity (Figure9A) and increased PU.1 protein using specific anti-PU.1 antibodies (Figure 9B).

Fig. 8.

Effect of DMSO-induced differentiation of HL-60 cells onp40phox mRNA expression and promoter function.

(A) Northern blot analysis of differentiating HL-60 cells. Cells were harvested after treatment with 1.25% DMSO for the indicated times, and total RNA was isolated. RNA (10 μg) was separated on a 1% agarose denaturing gel, transferred to nitrocellulose, and probed with a32P-labeled p40phox cDNA probe. For a loading control, the blot was stripped and reprobed with labeled cDNA of human acidic ribosomal phosphoprotein 36B4. (B) Effect of differentiation on p40phox promoter function. HL-60 cells were transfected with pGL3-Basic, pGL3-p40-106, or pGL3-p40-106-PU.1Mt and were treated with DMSO for 38 hours before assay for luciferase activity. Data are the means (± SE) of 3 independent experiments.

Fig. 8.

Effect of DMSO-induced differentiation of HL-60 cells onp40phox mRNA expression and promoter function.

(A) Northern blot analysis of differentiating HL-60 cells. Cells were harvested after treatment with 1.25% DMSO for the indicated times, and total RNA was isolated. RNA (10 μg) was separated on a 1% agarose denaturing gel, transferred to nitrocellulose, and probed with a32P-labeled p40phox cDNA probe. For a loading control, the blot was stripped and reprobed with labeled cDNA of human acidic ribosomal phosphoprotein 36B4. (B) Effect of differentiation on p40phox promoter function. HL-60 cells were transfected with pGL3-Basic, pGL3-p40-106, or pGL3-p40-106-PU.1Mt and were treated with DMSO for 38 hours before assay for luciferase activity. Data are the means (± SE) of 3 independent experiments.

Close modal
Fig. 9.

Effect of DMSO-induced differentiation of HL-60 cells on levels of PU.1 protein.

(A) EMSA of DMSO-induced cells. HL-60 cells were incubated with or without 1.25% DMSO for 38 hours, and nuclear extracts were prepared. EMSA was carried out with 5 μg nuclear extracts using the32P-labeled p40phox PU.1a DNA probe. Specific PU.1-containing complexes are indicated by the arrow. (B) Immunoblot analysis of DMSO-induced cells. HL-60 cells were incubated with or without 1.25% DMSO for 38 hours, and equal amounts of protein from whole cell lysates were separated by SDS-PAGE and analyzed by immunoblotting with specific antibody to PU.1. The 42-kd form of the protein is indicated by the arrow. The slower-migrating band probably corresponds to a hyper-phosphorylated form of the protein.67 68 

Fig. 9.

Effect of DMSO-induced differentiation of HL-60 cells on levels of PU.1 protein.

(A) EMSA of DMSO-induced cells. HL-60 cells were incubated with or without 1.25% DMSO for 38 hours, and nuclear extracts were prepared. EMSA was carried out with 5 μg nuclear extracts using the32P-labeled p40phox PU.1a DNA probe. Specific PU.1-containing complexes are indicated by the arrow. (B) Immunoblot analysis of DMSO-induced cells. HL-60 cells were incubated with or without 1.25% DMSO for 38 hours, and equal amounts of protein from whole cell lysates were separated by SDS-PAGE and analyzed by immunoblotting with specific antibody to PU.1. The 42-kd form of the protein is indicated by the arrow. The slower-migrating band probably corresponds to a hyper-phosphorylated form of the protein.67 68 

Close modal

PU.1, the most divergent member of the ets family of transcription factors, is important in the early development of multiple hematopoietic progenitors. Targeted disruption of the PU.1 locus results in multilineage defects that affect B and T lymphocytes, monocytes, osteoclasts, alveolar macrophages, and neutrophils.39,40 The PU.1 transcription factor is expressed specifically in hematopoietic tissues, particularly in cells of granulocytic, monocytic, and B lymphoid lineages.42,51-53 These patterns of expression are shared with the components of the phagocyte NADPH oxidase.1-3Moreover, previous studies have shown that the components of the phagocytic respiratory burst oxidase are transcriptionally regulated by PU.1.41,54-57 For example, expression of the large subunit of the membrane-bound cytochrome b558, gp91phox is controlled by PU.1, along with other transcription factors.54-57 Of particular note, the PU.1-binding site is critical for function of the promoter of thegp91phox (CYBB) gene, and point mutations in this site lead to the CGD clinical phenotype.55,56 In the case of the cytosolic oxidase component p47phox, we have shown that its myeloid-specific expression is regulated primarily through a single essential cis element that binds PU.1 with high avidity.41 In addition, we and others have reported that p67phox transcription is regulated by a complex array of transcription factors that includes PU.1.57-59Adding to the body of evidence for a key role of PU.1 in regulation of NADPH oxidase expression are our current data that the cytosolic oxidase component p40phox is also regulated by PU.1. We have shown that a unique combination of 3 PU.1-binding elements, 2 in the proximal promoter and one in the 5′ UTR of the gene, comprise the major regulatory components ofp40phox gene transcription in the HL-60 myeloid and Raji B-cell lines. Moreover, we have demonstrated that the 3 PU.1-binding sites also mediate increased expression of thep40phox gene observed during DMSO-induced granulocytic differentiation of HL-60 cells.

Recent studies indicate that regulation by multiple PU.1 promoter elements may not be unusual for myeloid-restricted genes. Two functional PU.1 sites have been identified in the promoter of the myeloid-specific CD18 gene60 and, interestingly, in the promoter of the PU.1 gene itself.61The PU.1-binding sites in the PU.1 promoter thus appear to act as a pathway for autoregulation, a potentially important concept for lineage development of myeloid cells. A recent report has indicated that a low level of expression of PU.1 in hematopoietic progenitors is associated with the development of B cells, whereas a high level of expression blocks B-cell development and promotes macrophage differentiation.62 Nevertheless, the specific functional implications of the presence of multiple functional PU.1 sites in a promoter are yet to be determined. It is possible that in some genes they could act in a synergistic or cooperative manner, allowing a relatively strong transcriptional response in the presence of low activity of the PU.1 transcription factor, a situation that might be found in the early stages of myeloid development. This may well be the case with the p40phox gene. Transcripts for p40phox can be readily identified in undifferentiated myeloid cell lines by Northern blot analysis,36 whereas the mRNA forp47phox, p67phox, andgp91phox is generally more difficult to detect.36,50,63 64 

We showed previously that the avidity of binding of PU.1 by a particular cis element correlates with its capacity to dictate reporter gene transcription.46 The results of our analyses of the 3 PU.1 sites in thep40phox promoter support this earlier observation. The affinity of binding of PU.1 to thep40phox PU.1 oligonucleotide probes in the gel mobility shift assay and the transacting activity mediated by each site in the transfection studies were of the same rank order (PU.1a is greater than PU.1b is greater than PU.1c). The relatively high activity of the PU.1a site is probably caused by the presence of optimal (AT-rich) sequences flanking the 5′ end of the core GAGGAA sequence.46 Interestingly, these sequences are not found in the weakly acting PU.1c site. The proximity of the 3 PU.1-binding elements to each other and their different activities suggest that together they produce a synergistic activation ofp40phox gene transcription. However, our deletion–mutation studies indicated that the relative contribution to promoter activity of each of the 3 PU.1 sites was not reduced when the other sites were functionally lost. Therefore, at least in resting HL-60 cells, it appears more likely that each PU.1 site acts independently of the others.

Transfection of the −1599 bp p40phoxpromoter construct into the nonmyeloid HeLa cell line resulted in moderate levels of reporter gene activity. Consistent with the absence of PU.1 protein in these cells, this activity was not affected by mutation of the PU.1 sites in the promoter construct. These data suggest that nonmyeloid-specific factors are capable of binding and transactivating the −1599 bp p40phoxpromoter construct in locations other than the PU.1 sites we have described here. However, if present in myeloid cells and B lymphocytes, these factors must be dependent on PU.1 binding because mutation of the PU.1 sites alone markedly reduced promoter activity in these cells. Given that the expression of p40phox is restricted to myeloid cells and B lymphocytes, transactivation of the gene by these putative factors in nonmyeloid tissues must be inhibited in some way. However, a previous report suggests that some myeloid-specific promoter–reporter constructs may show activity in HeLa cells, as opposed to other nonmyeloid cell types.65 

There is increasing evidence that PU.1 can act in association with accessory factors to transactivate genes. Early studies showed an essential functional requirement for the factor NF-EM5 (PIP) to act in conjunction with PU.1 to transactivate the immunoglobulin kappa gene.66 The recruitment of NF-EM5 to the PU.1–DNA complex was dependent on the activation of PU.1 by phosphorylation of serine 148, possibly by casein kinase II. In the immunoglobulin kappa gene, nonphosphorylated PU.1 was able to bind its cognate site but could not stimulate promoter activity. It is possible that the binding of PU.1 to the p40phox gene recruits such accessory factors into a ternary complex that in turn dictates promoter activity. Our data do not directly support such a model because the pattern of complexes formed on EMSA with thep40phox PU.1 probes was similar for HL-60 nuclear extracts and in vitro–synthesized PU.1 protein. However, the recruitment of accessory factors may require additional nucleotide sequences that extend beyond those used for the probes.

The induction of granulocytic differentiation in HL-60 cells with DMSO led to an increase in the net cellular levels ofp40phox mRNA. DMSO treatment also produced an increase in the total cellular levels of PU.1, particularly in the slower-migrating form of PU.1, which most likely corresponds to a hyper-phosphorylated form of the protein.67,68 These data are consistent with earlier studies using enriched human CD34+ progenitor cells in which the induction of myeloid differentiation with granulocyte macrophage–colony-stimulating factor resulted in an increase in PU.1 mRNA and protein.69 70 The increase in PU.1 protein in the HL-60 cells was accompanied by an increase in the levels of PU.1 binding to DNA and PU.1-mediatedp40phox promoter activity. Taken together with our functional mutation studies, these data suggest that the increased p40phox gene transcription observed during maturation and differentiation is mediated primarily by the 3 identified PU.1 sites. This does not preclude the contribution of additional accessory or coactivator factors, perhaps recruited through the activation and phosphorylation of PU.1. However, these putative factors would have to function through the PU.1 elements because mutation of these sites reduced the promoter activity in the HL-60 cells to near-background levels.

We thank Dr Michael J. Klemsz for providing PU.1 expression plasmids and Dr Long Wang for assistance with graphics and statistical analyses.

Supported by grant AI20866 from the National Institutes of Health (R.A.C.) and by a grant from the Research Resources Program of the Howard Hughes Medical Institute (S.-L.L.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Babior
BM
NADPH oxidase: an update.
Blood.
93
1999
1464
1476
2
Clark
RA
Activation of the neutrophil respiratory burst oxidase.
J Infect Dis.
179
1999
S309
S317
3
Segal
BH
Leto
TL
Gallin
JI
Malech
HL
Holland
SM
Genetic, biochemical, and clinical features of chronic granulomatous disease.
Medicine.
79
2000
170
200
4
Klebanoff
SJ
Oxygen metabolism and the toxic properties of phagocytes.
Ann Intern Med.
93
1980
480
489
5
Clark
RA
Extracellular effects of the myeloperoxidase-hydrogen peroxide-halide system.
Advances in Inflammation Research.
Weissmann
G
1983
107
146
Raven Press
New York, NY
6
Weiss
SJ
Tissue destruction by neutrophils.
N Engl J Med.
320
1989
365
376
7
Leusen
JHW
Verhoeven
AJ
Roos
D
Interactions between the components of the human NADPH oxidase: intrigues in the phox family.
J Lab Clin Med.
128
1996
461
476
8
DeLeo
FR
Quinn
MT
Assembly of the phagocyte NADPH oxidase: molecular interaction of oxidase proteins.
J Leukoc Biol.
60
1996
677
691
9
Volpp
BD
Nauseef
WM
Clark
RA
Two cytosolic neutrophil oxidase components absent in autosomal chronic granulomatous disease.
Science.
242
1988
1295
1297
10
Nunoi
H
Rotrosen
D
Gallin
JI
Malech
HL
Two forms of autosomal chronic granulomatous disease lack distinct neutrophil cytosol factors.
Science.
242
1988
1298
1301
11
Volpp
BD
Nauseef
WM
Donelson
JE
Moser
DR
Clark
RA
Cloning of the cDNA and functional expression of the 47 kilodalton cytosolic component of the human neutrophil respiratory burst oxidase.
Proc Natl Acad Sci USA.
86
1989
7195
7199
12
Abo
A
Pick
E
Hall
A
Totty
N
Teahan
CG
Segal
AW
Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1.
Nature.
353
1991
668
670
13
Rotrosen
D
Yeung
CL
Leto
TL
Malech
HL
Kwong
CH
Cytochrome b558: the flavin-binding component of the phagocyte NADPH oxidase.
Science.
256
1992
1459
1462
14
Segal
AW
West
I
Wientjes
F
et al
Cytochrome b-245 is a flavocytochrome containing FAD and the NADPH-binding site of the microbicidal oxidase of phagocytes.
Biochem J.
284
1992
781
788
15
Knaus
UG
Heyworth
PG
Kinsella
BT
Curnutte
JT
Bokoch
GM
Purification and characterization of Rac 2: a cytosolic GTP-binding protein that regulates human neutrophil NADPH oxidase.
J Biol Chem.
267
1992
23575
23582
16
Volpp
BD
Lin
Y
In vitro molecular reconstitution of the respiratory burst in B lymphoblasts from p47-phox–deficient chronic granulomatous disease.
J Clin Invest.
91
1993
201
207
17
Sekhsaria
S
Gallin
JI
Linton
GF
Mallory
RM
Mulligan
RC
Malech
HL
Peripheral blood progenitors as a target for genetic correction of p47 phox-deficient chronic granulomatous disease.
Proc Natl Acad Sci U S A.
90
1993
7446
7450
18
Li
F
Linton
GF
Sekhsaria
S
et al
CD34+ peripheral blood progenitors as a target for genetic correction of the two flavocytochrome b558 defective forms of chronic granulomatous disease.
Blood.
84
1994
53
58
19
Kume
A
Dinauer
MC
Retrovirus-mediated reconstitution of respiratory burst activity in X-linked chronic granulomatous disease cells.
Blood.
84
1994
3311
3316
20
Porter
CD
Parkar
MH
Verhoeven
AJ
Levinsky
RJ
Collins
MKL
Kinnon
C
p22-phox-deficient chronic granulomatous disease: reconstitution by retrovirus-mediated expression and identification of a biosynthetic intermediate of gp91-phox.
Blood.
84
1994
2767
2775
21
Someya
A
Nagaoka
I
Yamashita
T
Purification of the 260 kDa cytosolic complex involved in the superoxide production of guinea pig neutrophils.
FEBS Lett.
330
1993
215
218
22
Wientjes
FB
Hsuan
JJ
Totty
NF
Segal
AW
p40phox, a third cytosolic component of the activation complex of the NADPH oxidase to contain src homology 3 domains.
Biochem J.
296
1993
557
561
23
Tsunawaki
S
Mizunari
H
Nagata
M
Tatsuzawa
O
Kuratsuji
T
A novel cytosolic component, p40phox, of respiratory burst oxidase associates with p67phox and is absent in patients with chronic granulomatous disease who lack p67phox.
Biochem Biophys Res Commun.
199
1994
1378
1387
24
Fuchs
A
Dagher
M-C
Vignais
PV
Mapping the domains of interaction of p40phox with both p47phox and p67phox of the neutrophil oxidase complex using the two-hybrid system.
J Biol Chem.
270
1995
5695
5697
25
Ito
T
Nakamura
R
Sumimoto
H
Takeshige
K
Sakaki
Y
An SH3 domain-mediated interaction between the phagocyte NADPH oxidase factors p40phox and p47phox.
FEBS Lett.
385
1996
229
232
26
Fuchs
A
Dagher
MC
Fauré
J
Vignais
PV
Topological organization of the cytosolic activating complex of the superoxide-generating NADPH-oxidase: pinpointing the sites of interaction between p47phox, p67phox and p40phox using the two-hybrid system.
Biochim Biophys Acta Mol Cell Res.
1312
1996
39
47
27
Wientjes
FB
Panayotou
G
Reeves
E
Segal
AW
Interactions between cytosolic components of the NADPH oxidase: p40phox interacts with both p67phox and p47phox.
Biochem J.
317
1996
919
924
28
Tsunawaki
S
Kagara
S
Yoshikawa
K
Yoshida
LS
Kuratsuji
T
Namiki
H
Involvement of p40phox in activation of phagocyte NADPH oxidase through association of its carboxyl-terminal, but not its amino-terminal, with p67phox.
J Exp Med.
184
1996
893
902
29
Sathyamoorthy
M
De Mendez
I
Adams
AG
Leto
TL
p40 phox down-regulates NADPH oxidase activity through interactions with its SH3 domain.
J Biol Chem.
272
1997
9141
9146
30
Wilson
L
Butcher
C
Finan
P
Kellie
S
SH3 domain-mediated interactions involving the phox components of the NADPH oxidase.
Inflamm Res.
46
1997
265
271
31
Rinckel
LA
Faris
SL
Hitt
ND
Kleinberg
ME
Rac1 disrupts p67phox/p40phox binding: a novel role for Rac in NADPH oxidase activation.
Biochem Biophys Res Commun.
263
1999
118
122
32
Cross
AR
p40phox participates in the activation of NADPH oxidase by increasing the affinity of p47phox for flavocytochrome b558.
Biochem J.
349
2000
113
117
33
Grogan
A
Reeves
E
Keep
N
et al
Cytosolic phox proteins interact with and regulate the assembly of coronin in neutrophils.
J Cell Sci.
110
1997
3071
3081
34
Fuchs
A
Bouin
AP
Rabilloud
T
Vignais
PV
The 40-kDa component of the phagocyte NADPH oxidase (p40phox) is phosphorylated during activation in differentiated HL60 cells.
Eur J Biochem.
249
1997
531
539
35
Bouin
AP
Grandvaux
N
Vignais
PV
Fuchs
A
p40phox is phosphorylated on threonine 154 and serine 315 during activation of the phagocyte NADPH oxidase: implication of a protein kinase C type kinase in the phosphorylation process.
J Biol Chem.
273
1998
30097
30103
36
Zhan
S
Vazquez
N
Wientjes
FB
et al
Genomic structure, chromosomal localization, start of transcription and tissue expression of the human p40-phox, a new component of the nicotinamide adenine dinucleotide phosphate-oxidase complex.
Blood.
88
1996
2714
2721
37
Mizuki
K
Kadomatsu
K
Hata
K
et al
Functional modules and expression of mouse p40phox and p67phox, SH3-domain-containing proteins, involved in the phagocyte NADPH oxidase complex.
Eur J Biochem.
251
1998
573
582
38
Simon
MC
Olson
M
Scott
E
Hack
A
Su
G
Singh
H
Terminal myeloid gene expression and differentiation requires the transcription factor PU.1.
Curr Top Microbiol Immunol.
211
1996
113
119
39
Scott
EW
Simon
MC
Anastasi
J
Singh
H
Requirement of transcription factor PU.1 in the development of multiple hematopoietic lineages.
Science.
265
1994
1573
1577
40
McKercher
SR
Torbett
BE
Anderson
KL
et al
Targeted disruption of the PU.1 gene results in multiple hematopoietic abnormalities.
EMBO J.
15
1996
5647
5658
41
Li
SL
Valente
AJ
Zhao
SJ
Clark
RA
PU.1 is essential for p47phox promoter activity in myeloid cells.
J Biol Chem.
272
1997
17802
17809
42
Klemsz
MJ
McKercher
SR
Celada
A
Van Beveren
C
Maki
RA
The macrophage and B cell-specific transcription factor PU.1 is related to the ets oncogene.
Cell.
61
1990
113
124
43
Himmelmann
A
Thevenin
C
Harrison
K
Kehrl
JH
Analysis of the Bruton's tyrosine kinase gene promoter reveals critical PU.1 and SP1 sites.
Blood.
87
1996
1036
1044
44
Borregaard
N
Heiple
JM
Simons
ER
Clark
RA
Subcellular localization of the b-cytochrome component of the human neutrophil microbicidal oxidase: translocation during activation.
J Cell Biol.
97
1983
52
61
45
Boyd
KE
Wells
J
Gutman
J
Bartley
SM
Farnham
PJ
c-Myc target gene specificity is determined by a post-DNA binding mechanism.
Proc Natl Acad Sci U S A.
95
1998
13887
13892
46
Li
SL
Schlegel
W
Valente
AJ
Clark
RA
Critical flanking sequences of PU.1 binding sites in myeloid-specific promoters.
J Biol Chem.
274
1999
32453
32460
47
Collins
SJ
Ruscetti
FW
Gallagher
RE
Gallo
RC
Normal functional characteristics of cultured human promyelocytic leukemia cells (HL-60) after induction of differentiation by dimethyl sulfoxide.
J Exp Med.
149
1979
969
974
48
Tsiftsoglou
AS
Wong
W
Hyman
R
Minden
M
Robinson
SH
Analysis of commitment of human leukemia HL-60 cells to terminal granulocytic maturation.
Cancer Res.
45
1985
2334
2339
49
Levy
R
Rotrosen
D
Nagauker
O
Leto
TL
Malech
HL
Induction of the respiratory burst in HL-60 cells: correlation of function and protein expression.
J Immunol.
145
1990
2595
2601
50
Hua
J
Hasebe
T
Someya
A
Nakamura
S
Sugimoto
K
Nagaoka
I
Evaluation of the expression of NADPH oxidase components during maturation of HL-60 cells to neutrophil lineage.
J Leukoc Biol.
68
2000
216
224
51
Goebl
MG
The PU.1 transcription factor is the product of the putative oncogene Spi-1.
Cell.
61
1990
1165
1166
52
Galson
DL
Hensold
JO
Bishop
TR
et al
Mouse beta-globin DNA-binding protein B1 is identical to a proto-oncogene, the transcription factor Spi-1/PU.1, and is restricted in expression to hematopoietic cells and the testis.
Mol Cell Biol.
13
1993
2929
2941
53
Hromas
R
Orazi
A
Neiman
RS
et al
Hematopoietic lineage- and stage-restricted expression of the ETS oncogene family member PU.1.
Blood.
82
1993
2998
3004
54
Eklund
EA
Jalava
A
Kakar
R
PU.1, interferon regulatory factor 1, and interferon consensus sequence-binding protein cooperate to increase gp91phox expression.
J Biol Chem.
273
1998
13957
13965
55
Suzuki
S
Kumatori
A
Haagen
IA
et al
PU.1 as an essential activator for the expression of gp91phox gene in human peripheral neutrophils, monocytes, and B lymphocytes.
Proc Natl Acad Sci U S A.
95
1998
6085
6090
56
Voo
KS
Skalnik
DG
Elf-1 and PU.1 induce expression of gp91phox via a promoter element mutated in a subset of chronic granulomatous disease patients.
Blood.
93
1999
3512
3520
57
Eklund
EA
Kakar
R
Recruitment of CREB-binding protein by PU.1, IFN-regulatory factor-1, and the IFN consensus sequence-binding protein is necessary for IFN-gamma-induced p67phox and gp91phox expression.
J Immunol.
163
1999
6095
6105
58
Li
SL
Valente
AJ
Wang
L
Gamez
MJ
Clark
RA
Transcriptional regulation of the p67phox gene: role of AP-1 in concert with myeloid-specific transcription factors.
J Biol Chem.
276
2001
39368
39378
59
Gauss
KA
Bunger
PL
Quinn
MT
AP-1 is essential for p67phox promoter activity.
J Leukoc Biol.
71
2002
163
172
60
Rosmarin
AG
Caprio
D
Levy
R
Simkevich
C
CD18 (α2 leukocyte integrin) promoter requires PU.1 transcription factor for myeloid activity.
Proc Natl Acad Sci U S A.
92
1995
801
805
61
Chen
H
Ray-Gallet
D
Zhang
P
et al
PU.1 (Spi-1) autoregulates its expression in myeloid cells.
Oncogene.
11
1995
1549
1560
62
DeKoter
RP
Singh
H
Regulation of B lymphocyte and macrophage development by graded expression of PU.1.
Science.
288
2000
1439
1441
63
Obermeier
H
Sellmayer
A
Danesch
U
Aepfelbacher
M
Cooperative effects of interferon-gamma on the induction of NADPH oxidase by retinoic acid or 1,25(OH)2-vitamin D3 in monocytic U937 cells.
Biochim Biophys Acta Mol Cell Res.
1269
1995
25
31
64
Yu
LX
Zhen
L
Dinauer
MC
Biosynthesis of the phagocyte NADPH oxidase cytochrome b558: role of heme incorporation and heterodimer formation in maturation and stability of gp91phox and p22phox subunits.
J Biol Chem.
272
1997
27288
27294
65
Moulton
KS
Semple
K
Wu
H
Glass
CK
Cell-specific expression of the macrophage scavenger receptor gene is dependent on PU.1 and a composite AP-1/ets motif.
Mol Cell Biol.
14
1994
4408
4418
66
Eisenbeis
CF
Singh
H
Storb
U
Pip, a novel IRF family member, is a lymphoid-specific, PU.1-dependent transcriptional activator.
Genes Dev.
9
1995
1377
1387
67
Mao
C
Ray-Gallet
D
Tavitian
A
Moreau-Gachelin
F
Differential phosphorylations of Spi-B and Spi-1 transcription factors.
Oncogene.
12
1996
863
873
68
Lodie
TA
Savedra
RJ
Golenbock
DT
Van Beveren
CP
Maki
RA
Fenton
MJ
Stimulation of macrophages by lipopolysaccharide alters the phosphorylation state, conformation, and function of PU.1 via activation of casein kinase II.
J Immunol.
158
1997
1848
1856
69
Cheng
T
Shen
H
Giokas
D
Gere
J
Tenen
DG
Scadden
DT
Temporal mapping of gene expression levels during the differentiation of individual primary hematopoietic cells.
Proc Natl Acad Sci U S A.
93
1996
13158
13163
70
Voso
MT
Burn
TC
Wulf
G
Lim
B
Leone
G
Tenen
DG
Inhibition of hematopoiesis by competitive binding of transcription factor PU.1.
Proc Natl Acad Sci U S A.
91
1994
7932
7936

Author notes

Robert A. Clark, Department of Medicine, University of Texas Health Science Center, 7703 Floyd Curl Dr, San Antonio, TX 78229-3900; e-mail: clarkra@uthscsa.edu.

Sign in via your Institution