Aging-related Deficiency of CD28 Expression in CD4+ T Cells Is Associated with the Loss of Gene-specific Nuclear Factor Binding Activity*

Abbe N. Vallejo, Achim R. NestelDagger , Michael Schirmer§, Cornelia M. Weyand, and Jörg J. Goronzy

From the Department of Immunology and the Division of Rheumatology, Department of Medicine, Mayo Clinic Foundation, Rochester, Minnesota 55905

    ABSTRACT
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Changes in T cell populations and concomitant perturbation of T cell effector functions have been postulated to account for many aging-related immune dysfunctions. Here, we report that high frequencies of CD28null CD4+ T cells were found in elderly individuals. Because deviations in the function of these unusual CD4+ T cells might be directly related to CD28 deficiency, we examined the molecular basis for the loss of CD28 expression in CD4+ T cells. In reporter gene bioassays, the minimal promoter of the CD28 gene was mapped to the proximal 400 base pairs (bp) of the 5' untranslated region. CD28 deficiency was associated with the loss of two noncompeting binding activities within a 67-bp segment of the minimal promoter. These binding activities were not competed by consensus Ets, Elk, or AP3 motifs that were found within the sequence stretch. The DNA-protein complexes were also not recognized by antibodies to Ets-related transcription factors. Furthermore, introduction of mutations into the 67-bp segment at positions corresponding to the two DNA-protein interaction sites, i.e. nucleotides spanning -206 to -179 and -171 to -148, resulted in the loss of specific nuclear factor binding activities and the abrogation of promoter activity. These observations implicate at least two regulatory motifs in the constitutive expression of CD28. The loss of binding activity of trans-acting factors specific for these sequences may contribute to the accumulation CD4+CD28null T cells during aging.

    INTRODUCTION
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Among the many consequences of aging is the progressive decline in protective immunity. Elderly individuals have increased susceptibility to infections and blunted responses to vaccines. The incidence of various forms of malignancies and the risk of developing autoimmune diseases also increases with age. Although the molecular processes leading to the aging of the immune system are not yet clear, there is compelling evidence that changes in T cell populations and concomitant changes in T cell effector functions may underlie many aging-related immune dysfunctions (1). Because of thymic involution from the onset of reproductive maturity, aging leads to the inability to generate new T cells. Thus, there is a progressive replacement of naive (CD45RA+) T cells with those having a memory (CD45RO+, CD29hi, and CD44hi) phenotype (2-5). Paradoxically, expansion of the memory T cell population is not associated with enhanced memory immune responses in the elderly. Although the reason for this phenomenon is not known, previous studies indicate that T cells undergo functional changes with aging. Defects in signal transduction and, consequently, decreased proliferation and interleukin (IL)1-2 production have been reported (6-11). Other reported aging-related changes in T cell function are the inability of T cells to support B cell proliferation and immunoglobulin production (1, 12). Thus, T cell-dependent B cell responses are impaired with aging as demonstrated by the marked reduction in the efficacy of vaccines in the elderly (13, 14).

In an attempt to identify mechanisms of impaired immune responses, we studied a cohort of elderly individuals. Phenotypic T cell profiles of these individuals indicate that a subgroup had accumulated CD4+ T cells deficient in the expression of CD28, a membrane glycoprotein typically found on CD4+ T cells that provides the requisite costimulatory signal for T cell activation (15, 16). In the elderly, these CD28null cells can comprise up to 45% of the total CD4+ T cell compartment. A similar CD4+ T cell subset is typically found among patients with rheumatoid arthritis. Analysis of the T cell receptor usage of these in vivo expanded CD4+CD28null T cells (17, 18) indicated that they are oligoclonal (19) and persist for many years (20).

We propose that the emergence of CD4+CD28null T cells may reflect an aging immune system. Consistent with this hypothesis are the features of these cells, namely, their in vivo clonality and longevity, memory phenotype, and unusual functional profiles, including defective B cell help activity (17-22). All of these characteristics are considered to be hallmarks of an aging immune system (1). Inasmuch as CD4+CD28null T cells appear to be a distinct subset of functional T cells in the elderly but not among younger individuals, further assessment of the immunobiology of these cells may provide insight into the cellular processes involved in immunosenescence.

Our interest in studying the biology of CD4+CD28null T cells in humans lies in the well documented role of CD28 in immune responses as the major costimulatory molecule required for T cell activation (15, 16). In the mouse, the CD28 molecule is expressed on virtually all CD4+ T cells and has been unequivocally shown to be required for the maintenance of T cell proliferation, IL-2 production, and the de novo synthesis of many T cell-specific genes. In the absence of CD28-mediated signals, T cell recognition of antigen results in anergy or the induction of apoptosis. Perhaps the most compelling evidence of the critical requirement for CD28 in T cell-mediated immunity is the CD28-deficient mouse, which exhibits various forms of immune dysfunctions (23-26).

Among the important issues related to the biology and emergence of the CD4+CD28null T cells is the molecular basis for the deficiency of CD28 expression. The genomic and cDNA structures of the gene have been reported previously (27, 28), but the structural and biochemical requirements for gene transcription remain to be elucidated. Therefore, we initiated studies to examine the structural requirements for the constitutive expression of CD28. It is important to note that although CD28 is constitutively expressed on CD4+ T cells, previous studies showed that the levels of its expression are not static. The level of CD28 expression has been shown to either transiently increase or decrease, depending on the stimulus (29, 30), implying that the levels of its expression profoundly influence its costimulatory function.

The observed deficiencies in cell surface expression of CD28 among CD4+ T cells is correlated with a lack of CD28 mRNA.23 Similar observations have also been reported for transformed CD4+CD28null T cells without apparent genomic lesions of CD28 itself (27). Therefore, we evaluated the hypothesis that CD28 deficiency in human CD4+ T cells is associated with a transcriptional block. In the present paper, bioassays were carried out to assess the transcriptional activity of the CD28 gene promoter in CD28-deficient cells. Additionally, studies were conducted to identify sequence motifs that may be associated with the differential surface expression of CD28 in primary human CD4+ T cells. Given the important role of CD28 in the immune response, elucidation of the biochemical and molecular basis for the modulation of its expression will facilitate our understanding of the biology of CD4+CD28null T cells. Because these unusual T cells appear to be a feature of the elderly, studies of the pathways controlling CD28 expression will provide insight on molecular mechanisms associated with the emergence of CD28null cells in the aging CD4 T cell compartment.

    MATERIALS AND METHODS
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Study Population-- A total of 60 Caucasians who did not have a personal or family history of autoimmune diseases were chosen. Mononuclear cells were isolated from heparinized peripheral blood by isopycnic centrifugation over Ficoll-Hypaque (Amersham Pharmacia Biotech). Cells were subjected to standard immunofluorescence staining with monoclonal antibodies to CD3, CD4, and CD28 (Becton Dickinson Immunocytometry Systems, San Jose, CA). Cells were fixed with 1% paraformaldehyde in phosphate-buffered saline, and the lymphocyte population was analyzed by flow cytometry using either a FACScan or FACSVantage cytometer (Becton Dickinson Immunocytometry Systems). All analyses of the staining profiles of lymphocytes were carried out using PC-LYSIS II software (Becton Dickinson Immunocytometry Systems).

The frequency patterns of CD28+ and CD28null cells among CD3+CD4+ lymphocytes were analyzed as a function of age. Statistical analyses of the frequencies were performed using the Mann-Whitney test (SYSTAT for Windows, SYSTAT, Inc., Evanston, IL).

Cell Culture-- The CD28null CD4+ and CD28+CD4+ T cell clones used in this study have been described previously (17, 21). They were propagated in RPMI 1640 medium (BioWhittaker, Walkersville, MD) containing 10% fetal calf serum (Summit Biotech, Ft. Collins, CO), 2 mM L-glutamine, 100 units/ml penicillin, and 100 µg/ml streptomycin sulfate (Life Technologies, Inc.). To enhance their expansion, cultures were also supplemented with recombinant human IL-2 (Genzyme Diagnostics, Cambridge, MA) to a final concentration of 20 units/ml and cocultured with 1 × 105 cells/ml of neuraminidase-treated gamma -irradiated (10,000 rad) Epstein-Barr virus-transformed B lymphoblastoid cells every 7-10 days. Cells were maintained in a humidified 37 °C/7.5% CO2 incubator. Cells were periodically examined for their expression of CD3, CD4, and CD28 by flow cytometry.

Cell lines used in the study were derived from selected individuals with high percentages (>= 20%) of CD28nullCD4+ T cells. CD3+CD4+ cells were isolated from peripheral lymphocytes by standard fluorescence-activated cell sorting procedures and propagated in vitro. The CD28null and the CD28+ sublines were generated from their respective parental CD4+ lines by at least two successive cell sorting procedures. Purity of the sublines for their CD28 phenotype was assessed periodically by flow cytometry. With these approaches, the parent cell lines maintained to date are 30-40% CD4+CD28null, whereas the CD28+ sublines are generally >= 98% pure, and the CD28null sublines are 100% CD28-negative.

Jurkat and HUT78 cells were obtained from ATCC (Rockville, MD). Both cell lines were maintained in antibiotic-free RPMI 1640 medium supplemented with 2 mM L-glutamine. Jurkat cells were grown in 5% fetal calf serum (Summit Biotech) and 5% newborn calf serum (Life Technologies), and HUT78 cells were grown in 10% fetal calf serum. Expansion of HUT78 was enhanced by the addition of recombinant human IL-2 to a final concentration of 5 units/ml. Both cell lines were also maintained at a cell density of 107 cells/ml while in culture. Cells were cultured in a humidified 37 °C/5% CO2 incubator. Under these culture conditions, high efficiencies of transfection (see below) have been achieved.

Isolation of the CD28 5'-Untranslated Region (UTR)-- The genomic template for the cloning of the CD28 gene 5'-UTR was obtained by a combined strategy (see Fig. 2) of restriction enzyme digestion, Southern blotting, and by polymerase chain reaction (PCR). Genomic DNA isolated from peripheral blood lymphocytes of a healthy donor was prepared using standard techniques and digested with a mixture of restriction enzymes containing HindIII, BglII, and ScaI. Restriction fragments were fractionated by agarose gel electrophoresis and blotted onto nylon membranes. The appropriate fragment was identified by hybridization with a synthetic 41-bp oligonucleotide probe (see Fig. 2B, probe 41) corresponding to a segment within the CD28 5'-UTR described previously (28). This 41-bp segment of the 5'-UTR contains the putative Ets1 and AP3 binding motifs. This combination of the two motifs within the 41-bp segment was found to be unique for the CD28 gene. Ets and AP3 motifs were determined by structural analysis of the published sequence of the CD28 5'-UTR (28) using the Map program in GCG software (Genetics Computer Group, University of Wisconsin, Madison, WI) interfaced with the transcription factor data base (NCBI, National Institutes of Health, Bethesda, MD).

The appropriate restriction fragment was purified and used as a template for the isolation of the 5'-UTR by PCR with gene-specific primers using the Expand PCR System (Boehringer Mannheim). All PCR primers used in this study contained an engineered 5' restriction enzyme site to facilitate subsequent cloning into plasmid vectors. Synthesis and purification of oligonucleotide probes and primers and the PCR conditions have been described (31).

The appropriate PCR amplification products were identified by the Southern hybridization procedure described above. These PCR products were digested with the appropriate restriction enzyme, purified by GeneClean (BIO 101, La Jolla, CA), and cloned into a cloning/expression plasmid (see below). Plasmids were introduced into Escherichia coli DH5alpha (Life Technologies, Inc.) by standard transformation and drug selection protocols. Recombinants were screened initially by colony lift hybridization using the 41-bp probe followed by the direct PCR of positively hybridizing colonies using the same amplification primers used in the initial PCR. Clones containing the CD28 5'-UTR were authenticated by sequencing.

Reporter Gene Bioassays-- For the present studies, a cloning/reporter plasmid was specifically created. The plasmid, pSVGFPAmpR, was a fusion of the appropriate BamHI-HindIII fragments from pGLPromoter (Promega, Madison, WI) and phGFP-S65T (CLONTECH). It contains the ampicillin resistance gene, f1ORI, a multiple cloning site, and the minimal SV40 promoter. The humanized green fluorescence protein (hGFP) cDNA sequence with the associated SV40 poly(A) site is located downstream of the minimal SV40 promoter. This plasmid was used as one of the control plasmids in transient transfection assays, as well as the cloning vector for the construction of a series of hGFP reporter plasmids containing fragments of the CD28 5'-UTR.

Truncation variants of the CD28 5'-UTR were generated by PCR and cloned into the BglII and HindIII sites of pSVGFPAmpR after excision of the minimal SV40 promoter. These plasmids were amplified in bacteria as described above. Sequencing authenticated all CD28 5' UTR reporters generated, and two independent clones for each truncation variant were chosen for subsequent bioassays. All plasmids used in reporter bioassays were purified by cesium chloride density centrifugation. The plasmid preparations were then dialyzed against 1× Tris-EDTA, desalted by column chromatography (NAP-10 columns, Amersham Pharmacia Biotech), and stored at -20 °C.

Reporter plasmids were transiently transfected into Jurkat and HUT78 cells by electroporation. 1 × 107 cells resuspended in 300 µl of drug-free culture medium were aliquoted to electroporation cuvettes (4-mm gap Cuvette Plus, BTX, San Diego, CA). 28 µg of each hGFP reporter, resuspended in 50 µl of serum-free drug-free RPMI medium, was added to the cell suspension. The cell-DNA mixture was incubated on ice for 10 min and then electrically pulsed at 350 V with a capacitance of 960 µF and resistance of 129 ohms using an electroporator (ElectroCell 600, BTX Electroporation Systems). Cuvettes were immediately transferred onto ice and incubated for 10 min. About 1 ml of culture medium was then added to the cuvettes to resuspend the cells and subsequently transferred to sterile 6-ml tubes. Culture medium was added to the tubes to a final volume of 3.5 ml, and the cells were incubated overnight in a tissue culture incubator. hGFP expression was determined by flow cytometry 20 h after transfection. The analyses were carried out by electronically gating on viable cells by standard propidium iodide staining. For each experiment, a total of 50,000 events were collected for quantitative analysis.

In experiments involving mitogens, transiently transfected cells were initially incubated in tissue culture medium for 4 h before the mitogens were added. Phorbol 12-myristyl 13-acetate (PMA) (Sigma Chemical Co.) was added to a final concentration of 50 ng/ml, and ionomycin (Sigma) was added to a final concentration of 10 nM. In initial experiments (data not shown), these concentrations of PMA and ionomycin were found to promote the proliferation of primary CD4+ T cells and IL-2 production by Jurkat cells under the culture conditions described above.

For each transfection experiment, two control transfections were carried out. One of the controls involved the transfection of pSVGFPAmpR that served as the basal/vector control. The other involved transfection of phGFP-S65T (CLONTECH), a hGFP reporter under the control of the CMV promoter. hGFP reporter gene bioassays conducted also involved the cotransfection of 2 µg of the luciferase plasmid pCMVLuc (32). This was done to normalize for transfection efficiency of each test hGFP reporter. Luciferase activities were determined by bioluminescence using a bioassay kit (Promega), and photoemissions were measured by a luminometer (Lumat LB9501, Berthold Analytical, Nashua, NH).

Electrophoretic Mobility Shift Assays (EMSAs)-- In the present work, EMSAs were conducted using nuclear extracts from both CD28+ and CD28null CD4+ T cell lines and clones described above. Additional extracts from Jurkat, HUT78, other non-T cell ATCC lines (HeLa, K562, RD, and U937), and the Epstein-Barr virus-transformed B lymphoblastoid cell line HT10 were also prepared. Binding probes used in these assays were as indicated (see "Results" and Fig. 4). In competitive EMSAs, competitors were added to the binding reaction at 5-300-fold excess concentrations relative to the binding probe. The conditions for the preparation of nuclear extracts, radiolabeling of oligonucleotide probes, binding reactions, nondenaturing polyacrylamide electrophoresis, and visualization of gel shifts have been described (33).

Mutational Analysis-- Results of EMSAs indicated that there are two sequence motifs in the CD28 promoter 5'-UTR that bound nuclear factors from CD28+, but not CD28null, CD4+ T cells (see "Results"). To further evaluate the role of these two motifs in the promoter activity of the 5'-UTR, site-specific mutations were introduced into reporter constructs containing the minimal CD28 promoter (see "Results" and Fig. 3). Cluster of overlapping mutations, as indicated, were introduced by the gene splicing by overlap extension technique described previously (31). For these studies, the mutants were cloned into the BglII-HindIII sites of the vector, pGLPromoter (Promega) after the excision of the minimal SV40 promoter. Two independent clones for each mutant were selected. The plasmids were purified by cesium chloride density centrifugation and used in transient transfection assays with Jurkat cells as described above. For each transfection experiment, the plasmid pCMVLuc (32) was used as positive control, and the pGLPromoter plasmid served as vector control. Transfection efficiency was normalized by the cotransfection of 2 µg of the beta -galactosidase reporter, pCMVbeta gal (32). Luciferase and beta -galactosidase activities were determined by bioluminescence using the Dual-Light Assay kit (Promega), and photoemissions were measured by a luminometer (Lumat LB9501).

Mutants that showed ablation of reporter activity were identified, and their corresponding sequences were synthesized. Such oligonucleotides were used in EMSAs either as binding probes or as competitors to the wild type sequence. Preparation of nuclear extracts, conditions of binding reactions, and resolution and visualization of gel shifts were as described above.

Sequence Mapping of the CD28 5'-UTR-- The cloned 600-bp segment of the CD28 5'-UTR was subjected to transcription factor mapping using the Map program in the GCG software interfaced with the transcription factor data base. Mapping algorithms were set at high stringency in that the relevant nuclear factor binding sites were identified with exact matches or with a single allowable mismatch.

    RESULTS
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Deficiency of CD28 Expression in Human CD4+ T Cells Is Correlated with Age-- Our previous studies indicated that CD4+CD28null T cells are extremely infrequent in the majority of healthy individuals; however, a subset of individuals showed increased frequencies (18). We examined an extended cohort of healthy Caucasian individuals to identify factors associated with the increased numbers of CD4+ CD28null cells. As shown in Fig. 1, the frequency of CD4+ CD28null T cells was significantly correlated with age. These cells were rarely found among individuals younger than 40 years. In contrast, many of the elderly have a marked increase in the frequency of CD4+CD28null T cells. In some elderly individuals, these cells comprised up to 45% of the total CD4+ T cells (Fig. 1, inset). It is important to note that there were also elderly individuals who had extremely low frequencies of these unusual cells (<1% of the total CD4+ T cells) and those who completely lacked them. Conversely, there were occasionally those younger than 40 years with high frequencies (>2%) of CD4+CD28null T cells. These results indicate that there are other age-independent factors influencing the presence of these cells in the peripheral blood.


View larger version (23K):
[in this window]
[in a new window]
 
Fig. 1.   Age dependence of CD4+ CD28null T cell frequencies. Peripheral blood mononuclear cells from healthy volunteers were stained for CD3, CD4, and CD28, and their levels of expression were determined by flow cytometry. The frequencies of CD4+CD28null T cells were plotted as a function of age. Data shown are median frequencies of CD4+CD28null T cells in three different age groups (<40 years, n = 16; 40-59 years, n = 15; >= 60 years, n = 29) within the 25th and 75th percentiles as box plots and within the 10th and 90th percentile as whiskers. The indicated p values were determined by the Mann-Whitney U test. Inset, scatter plot of the frequencies of CD4+CD28null T cells among donors older than 55 years.

The Promoter Activity of the CD28 5'-UTR Is Localized to the Proximal 400-bp Region-- The deficiency in the cell surface expression of CD28 in CD4+ T cells was previously shown to be due to the uniform lack of CD28 mRNA.2, 3 By standard reverse transcription-PCR techniques, none of the splice variants of CD28 mRNA (27, 28) were detectable. To test the hypothesis that a transcriptional block might account for the deficiency in CD28 expression, studies were initiated to isolate the 5'-UTR of the gene for functional assays. The genomic structure of the human CD28 gene has been described (28). The restriction map of the gene has also been reconstructed by the mapping of contiguous phage clones. However, the sequence encompassing the entire gene domain remains to be determined. The regions of the gene that have been sequenced include 760 bp of the 5'-UTR, exons 1-4, and parts of introns 1-3. Intronic sequences that have been determined are those that immediately flank the exons. Using this information, we conducted a limited restriction enzyme analysis to isolate the appropriate genomic template for the cloning of the CD28 5'-UTR. For these studies, HindIII, BglII, and ScaI enzyme digestion was performed. As indicated previously (28), ScaI and HindIII sites are located within introns 1 and 2, respectively (Fig. 2A). Thus, a mixture of these enzymes was predicted to yield restriction fragments containing the CD28 5'-UTR. By Southern hybridization using a 41-bp sequence (Fig. 2B), the 5' UTR was found to be localized within a approx 2.5 kb HindIII fragment (Fig. 2A). This fragment was within larger BglII and ScaI fragments because digestion of DNA with the all three enzymes or a combination of HindIII with either BglII or ScaI yielded hybridizing fragments of the same size as the HindIII digested DNA. These results indicated that ScaI, BglII, and HindIII sites are situated on either side of exon 1. Using the Map programs in the GCG software, none of these restriction sites were found within exon 1 or the 760-bp sequence of the 5'-UTR (data not shown).


View larger version (23K):
[in this window]
[in a new window]
 
Fig. 2.   Isolation of the 5'-UTR of the human CD28 gene. Genomic DNA was subjected to restriction enzyme digestion and Southern blotting to identify the appropriate restriction fragment containing the CD28 gene domain. A, based on the Southern blots and previous data (28), a diagrammatic restriction map containing the CD28 5'-UTR was constructed. As shown, HindIII, BglII, and ScaI sites are situated on either side of exon 1. None of these sites have been found in exon 1 or in the 760-bp sequence of the 5'-UTR as determined by mapping in GCG. Also indicated in the map are the previously identified ScaI and HindIII sites, the 22-kb intron 1, and exon 2. B, using PCR and gene-specific primers (indicated by arrows), the proximal 600-bp region of the CD28 5'-UTR has been successfully cloned. Also depicted is the location and sequence of the 41mer sequence (probe 41) used as the hybridization probe in Southern blots. Base numbering of the 5'-UTR begins immediately upstream from the translation start codon, as shown.

Based on the results of the Southern blotting experiments, the genomic template for the cloning of the 5'-UTR by PCR was isolated. Using gene-specific primers, the proximal 600-bp of the 5'-UTR have been cloned. Because mechanisms involved in the normal expression of CD28 are not yet known, the cloned proximal region of the 5'-UTR was subsequently used in reporter gene bioassays to examine its promoter activity in transiently transfected cells. The cloned 600-bp region of the 5'-UTR and its truncation variants were cloned upstream of the hGFP cDNA sequence to generate a panel of reporter plasmids (Fig. 3A). These were transiently transfected into Jurkat cells, which express high levels of CD28 (data not shown), and hGFP expression was subsequently measured. Results of experiments showed that only cells transfected with reporters containing the proximal 400-bp segment of the 5'-UTR (plasmid 42) had significant hGFP activities (Fig. 3B). Such reporter activities were comparable to those seen with cells transfected with hGFP reporters under the control of the CMV promoter. Cells transfected with reporters containing the full 600-bp 5'-UTR (plasmid 52) showed low to negligible reporter activities, equivalent to those seen with cells transfected with the reporters containing the minimal SV40 promoter, which lacks enhancer activity.


View larger version (36K):
[in this window]
[in a new window]
 
Fig. 3.   Assessment of the promoter activity of the CD28 5'-UTR. A, a panel of hGFP reporters containing truncation variants of the cloned 600-bp sequence of the 5'-UTR was generated by standard PCR and cloning procedures. B, two clones for each hGFP reporter plasmid were randomly picked and transiently transfected into Jurkat T cells. hGFP expression was measured by flow cytometry 20 h after transfection. C, similar transfection experiments were also conducted to examine the effects of mitogens on promoter activity. Transfectants were cultured in medium alone (gray bars) or in the presence of 50 ng/ml PMA and 10 nM ionomycin (black bars). D, experiments were conducted to compare the promoter activity of the constructs in the CD28hi Jurkat T cells and the CD28null HUT78 T cells. In all experiments, hGFP activities were normalized against luciferase activities by cotransfection with the pCMVLuc plasmid (32). Luciferase activity was measured by bioluminescence. Data depicted are the average hGFP activities of two separate transfections for each reporter clone. hGFP activities are expressed as arbitrary units relative to the hGFP activity of the CMV-driven positive control reporter phGFP-S65T (CMV), which is shown as 100 units of maximal activity. Also shown are hGFP activities of transfections with the minimal SV40 promoter-driven reporter pSVGFPAmpR (minSV). Data shown are representative of four to six independent experiments in each data panel.

Previous studies indicated that although CD28 is constitutively expressed, the surface levels of CD28 expression may be modulated by mitogens (30). Thus, hGFP reporter bioassays were conducted to examine whether such mitogen-induced modulation may occur at the level of the gene promoter. As depicted in Fig. 3C, addition of 50 ng/ml PMA and 10 nM ionomycin to transiently transfected cells elicited reductions in hGFP levels. These reductions were evident in cells transfected with reporters containing the proximal 400 bp of the 5'-UTR (plasmid 42). Addition of mitogens to cells transfected with the other reporter constructs did not affect hGFP levels. The hGFP activities of cells transfected with CMV-driven reporters were also not significantly affected by incubation with mitogens.

The Promoter Activity of the Proximal 400-bp Segment of the 5'-UTR Is Suppressed in CD28null T Cells-- As indicated earlier, the deficiency of CD28 expression among CD4+ T cells was associated with a uniform lack of CD28 mRNA.2, 3 This suggested that a transcriptional block may be responsible for the lack of cell surface expression. To test this hypothesis, reporter gene bioassays were conducted using the CD4+CD28null T cell line HUT78. As we have found among our T cell clones, the lack of CD28 expression in HUT78 also occurred at the mRNA level without any apparent lesions in the genomic sequence of the gene (27). Results of transient transfection experiments with hGFP reporters showed that the promoter activity of the 400-bp segment of the CD28 5'-UTR was suppressed in HUT78 cells (Fig. 3D). The hGFP activities of cells transfected with the CMV promoter-driven reporters were maintained as expected. The negligible hGFP activities of HUT78 cells transfected with the other reporters were not significantly different from similarly transfected Jurkat cells.

A Lack of DNA-Protein Complex Formation with a 67-bp Sequence of the 5'-UTR Is Associated with a CD28-deficient Phenotype-- The observation that the promoter activity of proximal 400 bp of the CD28 5'-UTR was suppressed in HUT78 suggested that deficiency in CD28 expression may be associated with the lack of a relevant trans-acting factor(s). To evaluate this hypothesis, experiments were conducted to determine whether segments of the 400-bp region exhibited differential in vitro binding activities for nuclear factors from untransformed CD28+ and CD28null CD4+ T cells. In initial EMSAs, a 41-bp sequence (referred to as probe 41) was used (Figs. 2B and 4). Results of binding assays showed differential binding activities of probe 41 (Fig. 5). Binding activities to the probe were consistently observed with extracts from untransformed CD4+CD28+ T cell lines or clones and from Jurkat cells. In contrast, extracts from CD4+CD28null T cells, either transformed or untransformed, showed negligible binding activities. This differential binding activity was shown to localize to the opposite ends of probe 41. EMSAs using segments of probe 41 (Fig. 4), namely, the 5' half (probe 41A) and the 3' half (probe 41B), also showed differential binding activities (factors A and B) that correlated with the CD28 phenotype (Fig. 5). In contrast, binding assays using a probe corresponding to the central segment (probe 41C) did not show any binding activity regardless of the extract used in the assay. Interestingly, the binding activities seen with the 3' half (probe 41B) showed DNA-protein complexes with gel mobilities that were very similar to those seen with the intact 41-bp sequence (probe 41, factor A). This is in marked contrast to the activities seen with the 5' half (probe 41, factor B) which showed faster gel mobilities. Binding assays involving the same nuclear extracts using probes for the transcription factor SP1 (33) showed little difference among the T cell lines and clones examined. Moreover, EMSAs conducted using extracts from non-T cell lines also showed variable binding profiles (data not shown) that were unlike those seen with T cells as depicted in Fig. 5. Among the non-T cells examined were HeLa (epithelioid carcinoma), K562 (erythroid cell line), U937 (promonocytic line), RD (rhabdomyosarcoma), and HT10 (Epstein-Barr virus-transformed B lymphoblastoid line). These observations indicated that the differential binding activities of the 41-bp sequence or its specific segments were clearly correlated with the CD28 phenotype of the cells.


View larger version (17K):
[in this window]
[in a new window]
 
Fig. 4.   Sequence of a 67-bp subsegment of the proximal 400-bp region of the CD28 5'-UTR. The sequence shown is an internal segment of the 5'-UTR spanning positions -220 to -153. It contains putative binding motifs for Elk1, Ets, and AP3 as identified by the Map computer program in GCG interfaced with the transcription factor data base. Also shown is the 41mer sequence (probe 41) that was originally used as the hybridization probe for the isolation of the 5'-UTR (Fig. 2). The individual binding probes used in the EMSAs are indicated. The 67-bp segment overlaps with the previously reported transcription initiation site (28). Base numbering of the 5'-UTR begins immediately upstream from the translation start codon, as shown.


View larger version (77K):
[in this window]
[in a new window]
 
Fig. 5.   Differential binding activities of the 67-bp sequence for nuclear extracts from CD28+ and CD28null CD4+ T cells. Nuclear extracts from various T cell lines and T cell clones were prepared and analyzed in EMSAs. The conditions of the binding reactions were as described previously (32). About 15 µg of nuclear extracts was used in all assays. The binding probes were as shown (Fig. 4). DNA-protein complexes were resolved by nondenaturing polyacrylamide gel electrophoresis using 6% gels. The gels were dried and exposed to autoradiographic film. As a system control, EMSAs were also carried out using SP1 sequences (33). Data shown are optical images of representative autoradiograms. Binding profiles shown were reproducible in at least three experiments for each of five independent preparations of nuclear extracts. Lane 1, parental CD4+ T cell line DH; lane 2, sorted CD28+ DH subline; lane 3, sorted CD28null DH subline; lane 4, CD4+CD28+ T cell clone P43; lanes 5 and 6, CD4+CD28null T cell clones K2 and K9, respectively; lane 7, Jurkat cells; lane 8, HUT78 cells.

The differences between the gel mobilities of DNA-protein complexes formed with probes 41A and 41B suggested that these were two distinct activities. Because these probes were of the same length, the observed binding activities likely represent two different DNA-binding protein complexes. However, this is confounded by the observation that DNA-protein complexes formed with the intact 41-bp sequence showed very similar mobilities with that of the 3' half (probe 41B). These suggested that there are two nonoverlapping sites localized to either side of the sequence. To confirm these findings, probes including nucleotide positions upstream of the 41-bp probe were used (Fig. 4). Experiments showed that the binding activity seen with the 5' half (probe 41A) extended to nucleotide positions flanking the 41-bp sequence, i.e. probes 4441A and 44 (Fig. 5). DNA-protein complexes seen with these probes showed gel mobilities that were very similar to those with the 5' half (probe 41A). Similarly, the binding activities of the 3' half (probe 41B) were indistinguishable from probes that extended to downstream immediately flanking nucleotides (probe 5152; data not shown). Thus, the 5' and 3' binding activities were indeed distinct from each other. These observations also indicated that the segment of the CD28 5'-UTR that shows differential binding activities for nuclear extracts from CD28+ and CD28null CD4+ T cells encompasses a 67-bp sequence. Incidentally, this 67-bp sequence overlaps with the presumed transcription initiation site previously reported by Lee et al. (28).

There Are Two Noncompeting DNA-Protein Interaction Sites within the 67-bp Segment of the 5'-UTR-- To further ascertain that the two binding activities within the 67-bp sequence defined by probes 41A and 41B were nonoverlapping, competitive EMSAs were conducted. The experiments showed that probe 41A activities were not reciprocally competed by the 41B activities (Fig. 6). The respective DNA-protein complexes seen with the 41A probe were very stable in that even a 300-fold excess of competitor probe 41B or the 41B-related probe 5152 (data not shown) did not abrogate binding activity. Conversely, 41B activities were not competed by high concentrations of probe 41A or the 41A-related probes 4441A and 44. As expected, competition assays involving related probes 41A, 4441A, and 44 effectively competed each other, just as the related probes 41B and 5152 were effective competitors (Fig. 6 and data not shown).


View larger version (77K):
[in this window]
[in a new window]
 
Fig. 6.   Localization of two noncompeting nuclear factor binding activities within the 67-bp segment of the 5'-UTR. Nuclear extracts from CD4+CD28+ T cells were prepared and used in competitive EMSAs. Conditions of the binding reactions were as in Fig. 5 with the addition of increasing concentrations (5-300-fold excess; SP1, 300-fold excess only) of the indicated competitor. Data shown were obtained from experiments with extracts from sorted CD4+CD28+ subline of the DH cell line (Fig. 5). Similar results were obtained using the CD4+CD28+ T cell clone P43 and Jurkat cells (data not shown). Binding probes and competitors used are indicated. The use of previously described Ets-like (36), Elk-like (37), and AP3-like (42) sequences as competitors did not inhibit the binding activities of the indicated CD28-derived sequences (data not shown). Data depicted were reproducible in two experiments for each of three independent nuclear extract preparations.

Additionally, these two nonoverlapping binding activities within the CD28 promoter were not competed by other sequences containing the binding motifs for known transcription factors (Fig. 6 and data not shown). Among the sequences examined were nuclear factor kappa B, STAT1alpha , IRF1, and SP1 (33).

As indicated in Fig. 4, the 67-bp sequence from the CD28 5'-UTR contained the binding motifs for the Elk1, Ets1, and AP3 transcription factors. Elk1 and Ets1 are structurally related proteins (34, 35) that have been shown to play an important role in the expression of T cell-specific genes (36-40). In contrast, AP3 and AP3-like factors are not known to mediate T cell-specific gene transcription, although they have been implicated in the differentiation of myeloid cells (41) and in the synthesis of an IgE-induced mast cell-derived cytokine (42).

To determine whether Ets-related factors contribute to the observed binding activities of the 67-bp sequence, supershift EMSAs were conducted. Results showed that antibodies to Ets1, Elk1, or Elf did not affect the gel mobilities of DNA-protein complexes seen with either the 41A/4441A probes or the 41B/41 probes (data not shown). Furthermore, EMSAs using probes containing previously characterized Ets-specific (36) and Elk-specific (37) sequences did not show any detectable binding activity (data not shown). These results were consistent with the earlier observation that EMSAs using probes containing the presumptive CD28 Ets site (the 41C probe) did not show any detectable binding activity (Fig. 5) regardless of the T cell-derived nuclear extract used in these studies.

To further assess whether AP3 or AP3-like factors are involved in the binding activities of the CD28 67-bp segment, reciprocal competition assays were performed with probes 41B and 5152 (Fig. 4), an AP3 consensus sequence, and an AP3-like sequence of a recently described mast cell-derived IgE-induced cytokine (42). Results showed that whereas the CD28-derived probes 41B and 5152 had significant binding activity (Figs. 5 and 6 and data not shown), neither the AP3 nor the AP3-like probes had detectable binding activities. Furthermore, these two probes did not compete with the activities of probes 41B and 5152 (data not shown).

The Two DNA-Protein Interaction Sites Are Situated Immediately Downstream from the CAAT-TATA Boxes-- Mapping analysis of the cloned 600-bp segment of the CD28 5'-UTR showed that the 67-bp segment containing the two nonoverlapping DNA-protein interaction sites lies downstream from the putative CAAT and TATA boxes (Fig. 7). As indicated by the results of gel shift assays (Figs. 5 and 6), these two sites, herein referred to as sites alpha  and beta , lie on opposite sides of an Ets1-like binding sequence. Site alpha  was the 5' site defined by probes 41A and 4441A, whereas site beta  was the 3' site defined by probes 41B and 5152. As indicated, sites alpha  and beta  overlap with Elk1- and AP3-like sequences, respectively. As shown by the gel shift assays, however, neither Ets1, Elk1, nor AP3 appeared to influence the observed binding activities of sites alpha  and beta  (see above and Figs. 5 and 6).


View larger version (17K):
[in this window]
[in a new window]
 
Fig. 7.   Transcription factor mapping of the CD28 5'-UTR. The cloned 600-bp segment of the CD28 5'-UTR was subjected to sequence mapping for DNA-protein interaction sites using the Map programs in the GCG software interfaced to the transcription factor data base. The diagram indicates the location of sequence motifs identified by exact homology or a single allowable mismatch to known transcription factor binding sequences. Also shown is the sequence stretch containing two motifs, referred to as sites alpha  and beta , that bound nuclear factors from extracts of CD28+ but not CD28null, CD4+ T cells (see "Results" and Figs. 4-6).

Along the 600-bp stretch, other sequence motifs were also found (Fig. 7). Downstream from sites alpha  and beta , there appeared to be two nuclear factor kappa B (NFkappa B) sites, an interferon response element (IRE), and a cAMP-responsive element (CREB). Upstream from the CAAT box were sequences with homology to AP1, AP2, and a retinoic acid response element (RARE). Whether or not these motifs play a role in the induction of the CD28 promoter remains to be examined. Curiously, the distal 120 bp of the cloned 600-bp region of the CD28 5'-UTR contained two motifs with homology to a consensus repressor element (RepE) previously found to block transcriptional activities of various gene promoters.

Mutations in the Two DNA-Protein Interaction Sites Abrogate Nuclear Factor Binding and Ablate Reporter Activity of the Minimal CD28 Promoter-- To evaluate the significance of the nuclear factor binding activities of sites alpha  and beta , a panel of reporter constructs containing mutations in these CD28 promoter motifs were generated. To identify the critical subsites for both motifs, a cluster of four nucleotide substitutions were introduced across the sequence stretch, thereby generating a series of overlapping mutants. As depicted in Fig. 8, the mutagenesis yielded motif variants that ablated reporter gene activities in that mutations in either site alpha  or site beta  independently elicited the inactivation of the minimal CD28 promoter. Mutations in site alpha  that resulted in the loss of promoter activity were within a cluster of seven centrally located nucleotide positions, i.e. defined by mutants M3 and M4 and spanning positions -194 to -186, whereas in site beta , the inactivating mutations clustered in four to six more distal positions, i.e. defined by mutants M9, M10, and M11 and spanning positions -161 to -154. Curiously, mutations located between the two sites (mutant M5) did not ablate reporter activities.


View larger version (25K):
[in this window]
[in a new window]
 
Fig. 8.   Inactivation of the minimal CD28 promoter by mutations of the DNA-protein interaction sites alpha  and beta . Clusters of nucleotide substitutions, as indicated, were introduced into the minimal CD28 promoter construct 42 shown in Fig. 3), cloned into a luciferase reporter vector, and used in transient transfection assays with Jurkat cells. Data shown are the mean (± S.E.) luciferase activities of three separate but concurrent transfections for each of two clones of the wild type (WT) or mutant (M1-M11) constructs. Luciferase activities are expressed as fold induction over the minimal SV40 promoter-driven control luciferase reporter pGLPromoter. The specific reporter activities were normalized against a cotransfected reporter, the CMV-driven beta -galactosidase plasmid pCMVbeta gal (32). For these experiments, a CMV-driven luciferase reporter, pCMVLuc, showed activities over 100-fold above the control, as expected (data not shown). Luciferase and beta -galactosidase activities were determined by bioluminescence. Data shown are representative of two independent experiments involving triplicate concurrent transfections.

It is important to note that the inactivating mutations within sites alpha  and beta  were situated at the 3'-flank of the overlapping Elk1 and AP3 sequences, respectively (Figs. 4, 7, and 8). These results, together with the inability of Elk1 and AP3 consensus sequences to compete specific nuclear factor binding (see above), confirm that neither factors were involved in the DNA-protein complex formation of sites alpha  and beta .

To determine whether the ablation of reporter activities by specific mutations in either site alpha  or beta  was correlated with the loss of DNA-protein complexes, EMSAs were carried out using the indicated mutant sequences as binding and competitor probes. As depicted in Fig. 9, none of the mutants showed significant nuclear factor binding activities, unlike their wild type counterparts. Furthermore, the mutants did not compete the wild type binding sequences even at a 300-fold excess of the concentration of the binding probe.


View larger version (65K):
[in this window]
[in a new window]
 
Fig. 9.   Mutations of sites alpha  and beta  abrogate specific nuclear factor binding activities. Sequence variants of sites alpha  and beta  that inactivated promoter activity (M3, M4, M10, and M11, shown in Fig. 8) were synthesized and used as binding probes and competitors to the wild type sequence (probes 4441A and 5152; see Fig. 4) in EMSAs. The binding profiles depicted were reproducible in two experiments involving nuclear extracts from two different CD4+CD28+ T cell lines. The EMSAs were carried out as in Figs. 5 and 6.

    DISCUSSION
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

Studies presented here address molecular mechanism(s) underlying the deficiency in CD28 expression of CD4+ T cells. CD4+ CD28null T cells are uncommon in healthy young individuals but emerge in the elderly. In some cases, these cells comprise up to 45% of the CD4+ T cell compartment. Given the critical role of CD28 in the immune response (15, 16, 23, 24), defective CD28 expression may contribute to T cell dysfunction characteristic of immunosenescence. Here we show that the lack of CD28 expression correlates with the absence of two trans-acting factor binding activities within the minimal CD28 promoter.

Accumulation of CD4+CD28null T Cells in an Aging Immune System-- The occurrence of CD4+CD28null T cells at unusually high frequencies among the elderly (Fig. 1) raises the issue of the relevance of these cells in immunity of the elderly. As has been well documented in both mice and humans, CD28 is the critical costimulatory molecule required for T cell activation, proliferation, and effector function (16). Its absence or interruption of its interaction with its ligands results in T cell anergy or the induction of apoptosis (15, 23, 25). In mice, the targeted deletion of CD28 resulted in a severely immunocompromised animal (24, 26). Thus, the observed accumulation of CD4+CD28null T cells in vivo raises the hypothesis that CD28 deficiency leads to deviations in T cell effector function and contributes to age-related immune dysfunctions.

As demonstrated previously, CD4+CD28null T cells are functional and are not anergic as might be expected (21). T cell receptor cross-linking either by immobilized anti-CD3 or soluble anti-CD3 and monocytes has been found to elicit proliferation. Under these stimulation conditions, these cells also produce high levels of cytokines, among which are IL-2, IL-4, and gamma -interferon. However, despite the ability of these unusual T cells to respond to T cell receptor-mediated stimuli, CD4+CD28null T cells fail to express CD40 ligand and are unable to support B cell proliferation and immunoglobulin production in vitro (22). This corroborates previous studies that demonstrated the requirement of CD28 in T cell-dependent B cell-mediated immune responses (43). Along these lines, we propose that accumulation of CD4+CD28null T cells with age may contribute to the well-documented phenomenon of humoral hyporesponsiveness among the elderly. As epidemiologic data indicate, low responsiveness to vaccination is not uncommon among elderly individuals. For instance, the effectiveness of influenza vaccine is markedly reduced in the elderly as compared with younger individuals (13, 14).

Delineation of cis-Acting Elements and Identification of Sequence Motifs Associated with CD28 Deficiency-- Inasmuch as the regulatory pathways involved in the normal expression of CD28 are not yet known, the present work provides basic information on the minimal structural requirements for CD28 gene transcription. Here, we have cloned 600 bp of the 5'-UTR and provide evidence that the proximal 400 bp contain the minimal cis-acting elements necessary for gene expression as assessed by reporter gene bioassays. As the data show, only reporter plasmids containing the proximal 400 bp yielded promoter activity. Structural analysis of the 5'-UTR (Fig. 7) revealed that this proximal 400-bp segment contained the putative CAAT and TATA boxes and various constituent binding sites where the RNA polymerase complex presumably assembles (44, 45). Deletion of this region, as shown by transfection experiments with reporters containing only the most proximal 200-bp segment (Fig. 3B), did not yield any significant promoter activity. Conversely, deletion of the proximal 200 bp also did not yield significant promoter activity, indicating that the TATA-containing region requires downstream elements to function.

Flanking the 400-bp region is a 200-bp segment that appeared to be inhibitory to promoter activity. The reason for this inhibition is not clear. However, results of the structural analysis (Fig. 7) revealed the presence of sequences within this distal 200-bp segment that are homologous to repressor elements found in other genes.

As shown previously, CD28 expression transiently increases upon T cell activation, and ligation of CD28 by specific antibody induces reduction in its surface expression (16, 29). These findings suggest that there are regulatory elements responsive to T cell activation signals that are involved in feedback control. Our data indicate that such elements may be contained within the proximal 400-bp minimal promoter. Cells transfected with reporter constructs under the control of this minimal 400-bp promoter showed significantly down-regulated reporter activity when exposed to ionomycin and PMA (Fig. 3C).

An important finding of the present study was that the promoter activity of the proximal 400-bp segment of the 5'-UTR is suppressed in HUT78 cells (Fig. 3D). This CD4+ T cell lymphoma has been shown to lack CD28 at both the mRNA and protein levels without apparent genomic lesions in the CD28 gene itself (27). This observation, together with the fact that reverse transcription-PCR analysis on a variety of untransformed CD4+CD28null T cell lines and T cell clones showed the uniform absence of CD28 transcripts,2, 3 supports the notion that deficiency in CD28 expression may be due to the lack of relevant trans-acting factors.

This hypothesis was evaluated by EMSAs with nuclear extracts from CD28+ and CD28null CD4+ T cells. The oligonucleotide probes used encompass a 67-bp segment containing putative binding sites for the Elk1, Ets1, and AP3 transcription factors (Figs. 4 and 7). As the data show, there are two noncompeting DNA binding activities within this sequence for extracts from CD28+ but not CD28null CD4+ T cells. These binding activities correspond to two nucleotide stretches that lie on opposite flanks of the Ets1 motif (Figs. 4-7). The 5' site, referred to as site alpha , is defined by probes 41A and 4441A and spans positions -206 to -179 of the minimal promoter. The 3' site, referred to as site beta , is defined by probes 41B and 5152 and spans positions -171 to -148 of the minimal promoter.

Although site alpha  overlaps with an Elk1, the observed binding activity does not appear to involve Elk- and Ets-related transcription factors as determined by three lines of evidence. First, there is a lack of any detectable binding activity with a subsite of the 67-bp sequence containing the Ets motif (Figs. 4 and 5, probe 41C). Moreover, similar assays using binding probes containing Ets- and Elk-specific sequences previously characterized for other genes showed negligible binding activities. Second, the observed binding activities are not recognized by antibodies to Ets1, Elk1, and Elf, the three predominant lymphoid cell-specific members of the Ets family of transcription factors (36, 46-50). Therefore, the direct binding of Ets-related factors to DNA does not account for the observed binding activities, but the possibility that Ets-related factors are a component of such activities cannot be totally ruled out. Previous studies show that Ets family members may form complexes with DNA-binding proteins (37, 51, 52). Hence, it is possible that such interactions can bury antibody epitopes. Third, results of the mutational analysis show that nucleotide positions that abrogate factor binding activity of site alpha  are situated 3' of the Elk1-like sequence (Figs. 8 and 9), strong evidence that the latter may not be involved in DNA-protein complex formation.

Site beta , on the other hand, overlaps with an AP3-like sequence. However, it is also unlikely that the observed differential binding activities of site beta  for nuclear extract from CD28+ and CD28null CD4+ T cells represent an AP3 or an AP3-like factor. AP3 has not been documented to play any role in T cell-specific gene transcription, although it has been shown to be important in the differentiation of myeloid cells (41). An AP3-like transcription factor has also been implicated in the inducible expression of a novel mast cell-derived cytokine (42). In the present study, known AP3-like sequences did not compete with the binding activities of the CD28 67-bp sequence. In a manner similar to that of site alpha , DNA-protein complex formation with site beta  is also abrogated by mutations situated 3' of the overlapping AP3-like sequence (Figs. 8 and 9), indicating that AP3-like factors may not be involved.

Taken together, the data presented are consistent with the hypothesis that the deficiency in CD28 expression in CD4+ T cells is associated with the lack of relevant trans-acting factors, among which are those specific for sites alpha  and beta . The observed binding activities of these two sites are clearly correlated with the CD28 phenotype and are uniformly found among CD4+ T cells. The loss of binding activities of sites alpha  and beta  due to mutations strongly correlates with the inactivation of the minimal CD28 gene promoter (Figs. 8 and 9), indicating that these two sites are critical to the initiation of transcription. The requirement of sites alpha  and beta  for promoter activity is further supported by the findings that neither the segment containing the CAAT and TATA boxes (-400 to -200 bp of the 5'-UTR) nor the proximal 200-bp region (containing sites alpha  and beta ) by themselves exhibit promoter activity (Figs. 3B and 7). These observations strongly indicate that the factors bound by sites alpha  and beta  are required for the initiation of transcription.

The 67-bp segment containing sites alpha  and beta  overlaps with the presumed transcription initiation site previously reported by Lee et al. (28). As noted by the authors, the reported length of the CD28 transcription initiation site is rather imprecise because of structural constraints brought about by upstream Alu sequences. Thus, the reported transcription initiation site is, at best, a partial map.

Regardless of the actual length of the transcription initiation site, sites alpha  and beta  are contained within this 67-bp region and are situated immediately downstream from the putative TATA box (Fig. 7). The significance of this unusual topographic arrangement is not known at this time, but at least two hypotheses may be postulated as to the role the two sites in CD28 gene transcription. One hypothesis is that sites alpha  and beta  may comprise the equivalent of an Initiator element (Inr) described in other genes (45, 53, 54). It is thought that such elements interact with TATA-bound complex and are critical in directing synthesis of initiated transcripts (45). Inrs may overlap with or encompass the entire transcription initiation site (45), as has been found in the promoters of eukaryotic initiation factor 2alpha , terminal deoxynucleotide transferase, dihydrofolate reductase, and adenovirus major late protein (54). The common feature of Inrs is that mutations can block transcriptional activation (45, 53, 54). In this regard, we show in the present work that mutations within sites alpha  and beta  do inactivate the minimal CD28 promoter (Fig. 8). A structural analysis of sites alpha  and beta  (data not shown), however, reveals the lack of sequence homology with the known families of Inr (54).

An alternative hypothesis is that sites alpha  and beta  bind factors that either are components of or are independent activators of the TATA-bound RNA polymerase complex. Although many of the core protein components of the TATA-bound complex have been described, it is possible that there are yet uncharacterized additional components (44, 45). It is conceivable that among these components are cell- or tissue-specific factors. Along these lines, we currently have evidence that sites alpha  and beta  binding activities are seen only with lymphoid tissues and with various T cell lines that express CD28 on their surface.4

Whether the site alpha - and beta -bound factors are initiators or gene-specific components of the TATA-bound protein assemblage, our data consistently demonstrated their critical role in the the promoter activity of the CD28 5'-UTR. The identification of the factors bound by the two sites may be among the key information that will further elucidate the biochemical pathway for CD28 gene transcription. Ultimately, the identification of the genes encoding for alpha - and beta -binding proteins will facilitate our understanding of the origin, fate, and biology of CD4+CD28null T cells found in elderly individuals.

    ACKNOWLEDGEMENTS

We thank Brenda Goehring for technical assistance and James Fullbright for assistance in statistical analysis and manuscript preparation.

    FOOTNOTES

* This work was supported by the Mayo Foundation, the Deutscher Akademischer Austauschdienst (Germany), Grants RO1 AR41974 and RO1 AR42527 from the National Institutes of Health, and Grant AF16 from the National Arthritis Foundation.The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Dagger Present address: School of Medicine, Ruprecht-Karls-Universität 69120 Heidelberg, Germany.

§ Present address: Department of Medicine, University of Innsbruck, A-6020 Innsbruck, Austria.

To whom correspondence should be addressed: Mayo Clinic Foundation, 200 1st St. S.W., Rochester, MN 55905. Tel: 507-284-1650; Fax: 507-284-5045.

1 The abbreviations used are: IL, interleukin; EMSA, electrophoretic mobility shift assay; hGFP, humanized green fluorescence protein; PCR, polymerase chain reaction; PMA, phorbol 12-myristyl 13-acetate; RA, rheumatoid arthritis; UTR, untranslated region; bp, base pair(s); GCG, Genetics Computer Group; CMV, cytomegalovirus.

2 J. J. Goronzy and C. M. Weyand, unpublished data.

3 T. Namekawa, C. M. Weyand, and J. J. Goronzy, submitted for publication.

4 A. N. Vallejo, J. C. Brandes, C. M. Weyand, and J. J. Guronzy, manuscript in preparation.

    REFERENCES
Top
Abstract
Introduction
Materials & Methods
Results
Discussion
References

  1. Miller, R. A. (1996) Science 273, 70-74[Abstract]
  2. Nagelkerken, L., Hertogh-Huijbregts, A., Dobber, R., and Drager, A. (1991) Eur. J. Immunol. 21, 273-281[Medline] [Order article via Infotrieve]
  3. Ernst, D. N., Weigle, W. O., Noonan, D. J., McQuitty, D. N., and Hobbs, M. V. (1993) J. Immunol. 151, 575-587[Abstract/Free Full Text]
  4. Gabriel, H., Schmitt, B., and Kindermann, W. (1993) Eur. J. Immunol. 23, 2704-2706[Medline] [Order article via Infotrieve]
  5. Dobber, R., Tielemans, M., Weerd, H. D., and Nagelkerken. (1994) Int. Immunol. 6, 1227-1234[Abstract]
  6. Ernst, D. N., Weigle, W. O., and Hobbs, M. V. (1993) Stem Cells 11, 487-498[Abstract]
  7. Hobbs, V. M., Weigle, W. O., Noonan, D. J., Torbett, B. E., McEvilly, R. J., Koch, R. J., Cardenas, G. J., and Ernst, D. N. (1993) J. Immunol. 150, 3602-3614[Abstract/Free Full Text]
  8. Song, L., Kim, Y. H., Chopra, R. K., Proust, J. J., Nagel, J. E., Nordin, A. A., and Adler, W. H. (1993) Exp. Gerontol. 28, 313-321[CrossRef][Medline] [Order article via Infotrieve]
  9. Shi, J., and Miller, R. A. (1993) J. Immunol. 151, 730-739[Abstract/Free Full Text]
  10. Utsuyama, M., Varga, Z., Fukami, K., Homma, Y., Takenawa, T., and Hirokawa, K. (1993) Int. Immunol. 5, 1177-1182[Abstract]
  11. Tu, K. Y., Matthews, R., and Matthews, K. S. (1995) Mech. Ageing Dev. 82, 105-128[CrossRef][Medline] [Order article via Infotrieve]
  12. Yang, X., Stedra, J., and Cerny, J. (1996) J. Exp. Med. 183, 959-970[Abstract]
  13. Fagiolo, U., Amadori, A, Cozzi, E., Bendo, R., Lama, M., Douglas, A., and Palu, G. (1993) Aging 5, 451-458[Medline] [Order article via Infotrieve]
  14. Degelau, J. J., O'Leary, J. J., and Hallgren, H. M. (1994) Aging: Immunol. Infect. Dis. 5, 27-41
  15. Linsley, P., and Ledbetter, J. (1993) Annu. Rev. Immunol. 11, 191-212[CrossRef][Medline] [Order article via Infotrieve]
  16. Lenschow, D. J., Walunas, T. L., and Bluestone, J. A. (1996) Annu. Rev. Immunol. 14, 233-258[CrossRef][Medline] [Order article via Infotrieve]
  17. Schmidt, D., Goronzy, J. J., and Weyand, C. M. (1996) J. Clin. Invest. 97, 2027-2037[Abstract/Free Full Text]
  18. Martens, P. B., Goronzy, J. J., Schaid, D., and Weyand, C. M. (1997) Arthritis Rheum. 40, 1106-1114[Medline] [Order article via Infotrieve]
  19. Schmidt, D., Martens, P. B., Weyand, C. M., and Goronzy, J. J. (1996) Molec. Med. 2, 608-618[Medline] [Order article via Infotrieve]
  20. Waase, I., Kayser, C., Carson, P. J., Goronzy, J. J., and Weyand, C. M. (1996) Arthritis Rheum. 39, 904-913[Medline] [Order article via Infotrieve]
  21. Park, W., Weyand, C. M., Schmidt, D., and Goronzy, J. J. (1997) Eur. J. Immunol. 27, 1082-1090[Medline] [Order article via Infotrieve]
  22. Weyand, C. M., Brandes, J. C., Schmidt, D., and Goronzy, J. J. (1998) Mech. Ageing Dev., in press
  23. Harding, F. A., McArthur, J. G., Gross, J. A., Raulet, D. H., and Allison, J. P. (1992) Nature 356, 607-610[CrossRef][Medline] [Order article via Infotrieve]
  24. Shaninian, A., Pfeffer, K., Lee, K. P., Kundig, T. M., Kishihara, K., Wakeham, A., Kawai, K., Ohashi, P. S., Thompson, C. B., and Mak, T. W. (1993) Science 261, 609-612[Medline] [Order article via Infotrieve]
  25. Corry, D. B., Reiner, S. L., Linsley, P. S., and Locksley, R. M. (1994) J. Immunol. 153, 4142-4148[Abstract/Free Full Text]
  26. Boise, L. H., Noel, P. J., and Thompson, C. B. (1995) Curr. Opin. Immunol. 7, 620-625[CrossRef][Medline] [Order article via Infotrieve]
  27. Aruffo, A., and Seed, B. (1987) Proc. Natl. Acad. Sci. U. S. A. 84, 8573-8577[Abstract]
  28. Lee, K. P., Taylor, C., Petryniak, B., Turka, L. A., June, C. H., and Thompson, C. B. (1990) J. Immunol. 145, 344-352[Abstract/Free Full Text]
  29. Turka, L. A., Ledbetter, J. A., Lee, K., June, C. H., and Thompson, C. B. (1990) J. Immunol. 144, 1646-1653[Abstract/Free Full Text]
  30. Linsley, P. S., Bradshaw, J., Urnes, M., Grossmaire, L., and Ledbetter, J. A. (1993) J. Immunol. 150, 3161-3169[Abstract/Free Full Text]
  31. Vallejo, A. N., Pogulis, R. J., and Pease, L. R. (1994) PCR Methods Appl. 4, S123-S130[Medline] [Order article via Infotrieve]
  32. Vallejo, A. N., Allen, K. S., and Pease, L. R. (1995) Int. Immunol. 7, 853-859[Abstract]
  33. Vallejo, A. N., and Pease, L. R. (1996) J. Biol. Chem. 271, 29813-29821[Abstract/Free Full Text]
  34. MacLeod, L., K., Leprince, D., and Stehelin, D. (1992) Trends Biochem. Sci. 17, 251-256[CrossRef][Medline] [Order article via Infotrieve]
  35. Seth, A., Ascione, R., Fisher, R. J., Mavrothalassitis, G. J., Bhat, N. K., and Papas, T. S. (1992) Cell Growth Differ. 3, 327-334[Medline] [Order article via Infotrieve]
  36. Ho, I. C., Bhat, N. K., Gottschalk, L. R., Lindsten, T., Thompson, C. B., Papas, T. S., and Leiden, J. M. (1990) Science 250, 814-818[Medline] [Order article via Infotrieve]
  37. Hipskind, R. A., Rao, V. N., Mueller, C. G. F, Reddy, E. S. P., and Nordheim, A. (1991) Nature 354, 531-534[CrossRef][Medline] [Order article via Infotrieve]
  38. Rao, V. N., and Reddy, E. S. (1992) Oncogene 7, 65-70[Medline] [Order article via Infotrieve]
  39. Leung, S., McCracken, S., Ghysdael, and Miyamoto, N. G. (1992) Oncogene 8, 989-997
  40. Salmon, P., Giovane, A., Wasylyk, B., and Klatzmann, D. (1993) Proc. Natl. Acad. Sci. U. S. A. 90, 7739-7743[Abstract/Free Full Text]
  41. Adler, V., and Kraft, A. S. (1995) J. Cell. Physiol. 164, 26-34[Medline] [Order article via Infotrieve]
  42. Jarmin, D. I., Kulmburg, P. A., Huber, N. E., Baumann, G., Prieschl-Strassmayr, E. E., and Baumruker, T. (1994) J. Immunol. 153, 5720-5729[Abstract/Free Full Text]
  43. Hathcock, K. S., and Hodes, R. J. (1996) Adv. Immunol. 62, 131-166[Medline] [Order article via Infotrieve]
  44. Zamel, L., and Reinberg, D. (1995) Annu. Rev. Biochem. 64, 533-561[CrossRef][Medline] [Order article via Infotrieve]
  45. Goodrich, J. A., Cutler, G., and Tjian, R. (1996) Cell 84, 825-830[Medline] [Order article via Infotrieve]
  46. Bhat, N. K., Thompson, C. B., Lindsten, T., June, C. H., Fujiwara, S., Koizumi, S., Fisher, R. J., and Papas, T. S. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 3723-3727[Abstract]
  47. Leiden, J. M., Yang, C. Y., Petryniak, B., Markovitz, D. M., Nabel, G. J., and Thompson, C. B. (1992) J. Virol. 66, 5890-5897[Abstract]
  48. Thompson, C. B., Wang, C. Y., Ho, I. C., Bohajanen, P. R., Petryniak, B., Hune, C. H., Miesfeldt, S., Zhang, L., Nabel, G. J., Karpinski, B., and Leiden, J. M. (1992) Mol. Cell. Biol. 12, 1043-1053[Abstract]
  49. Wurster, A. L., Siu, G., Leiden, J. M., and Hedrick, S. M. (1994) Mol. Cell. Biol. 14, 6452-6463[Abstract]
  50. Romano-Spica, V., Georgiou, P., Suzuki, H., Papas, T. S., and Bhat, N. K. (1995) J. Immunol. 154, 2724-2732[Abstract/Free Full Text]
  51. Bassuk, A. G., and Leiden, J. M. (1995) Immunity 3, 223-237[Medline] [Order article via Infotrieve]
  52. Wasylyk, B., Wasylyk, C., Flores, P., Begue, A., Leprince, D., and Stehelin, D. (1990) Nature 346, 191-193[CrossRef][Medline] [Order article via Infotrieve]
  53. Smale, S. T., and Baltimore, D. (1989) Cell 57, 103-113[Medline] [Order article via Infotrieve]
  54. Weis, L., and Reinberg, D. (1992) FASEB J. 6, 3300-3309[Abstract/Free Full Text]


Copyright © 1998 by The American Society for Biochemistry and Molecular Biology, Inc.