Interleukin-15 (IL-15) is an important lymphokine regulating natural killer (NK) activity, T-cell proliferation, and T-cell cytotoxic activities. We hypothesized that the reduced expression and production of IL-15 from cord blood (CB) may contribute to the immaturity of CB immunity and potentially delay immune reconstitution after CB transplantation. We compared the expression and production of IL-15 from activated cord versus adult mononuclear cells (MNCs) and the regulatory mechanisms associated with IL-15 expression in CB MNCs. We have also studied the effect of exogenous IL-15 stimulation on CB and adult peripheral blood (APB) MNCs in terms of NK and lymphokine-activated killer (LAK) activities and cytokine induction. Lipopolysaccharide (LPS)-stimulated CB and APB MNCs were used to determine IL-15 expression and protein production by Northern analysis and Western immunoblot analysis. IL-15 mRNA expression and protein accumulation in CB MNC were 25% ± 2.0% (12 hours, n = 4, P < .05) and 30% ± 2.5% (12 hours, n = 3, P < .05), respectively, when compared with APB MNCs. Nuclear run-on assays showed no differences between CB and APB MNCs during basal levels of transcription and after transcriptional activation. However, the half-life of IL-15 mRNA was approximately twofold lower in activated CB MNCs than in activated APB MNCs (CB: 101 ± 5.8 minutes v APB: 210 ± 8.2 minutes, n = 3, P < .05). Exogenous IL-15 significantly enhanced CB NK and LAK activities up to comparable levels of APB (P < .05). IL-15 also significantly induced interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) protein production (days 1, 3, and 6, P < .05, n = 3) in CB MNCs. IL-15–stimulated LAK cells induced a significant lytic response against two acute lymphoblastic cell lines and two pediatric neuroblastoma cell lines. Both NK and LAK activities were augmented by the combination of IL-12 and IL-15, and the low-dose combination of IL-12 and IL-15 achieved similar levels of in vitro NK and LAK cytotoxicity compared with higher doses of either lymphokine. The present study suggests that IL-15 mRNA and protein expression is decreased in activated CB, secondary, in part, to altered posttranscriptional regulation. The reduced production of IL-15 from CB MNCs in response to stimulation may contribute to the decrease in IFN-γ and TNF-α production and CB cellular immunity. However, exogenous IL-15 enhanced IFN-γ and TNF-α production and NK and LAK cytotoxicities in CB MNCs. The reduced production of IL-15 from activated CB may contribute to the immaturity of CB cellular immunity and delayed immune reconstitution after unrelated CB transplantation. Exogenous IL-15 administration may compensate for the immaturity of CB immunity. The synergistic in vitro effects of low-dose IL-12 and IL-15 also implies the possible use of low doses each of IL-12 and IL-15 for enhancing immune reconstitution and/or possibly as a form of antitumor immunotherapy after CB transplantation.

WE HAVE RECENTLY shown that umbilical cord blood (CB) could be used successfully as an alternative source of hematopoietic stem cells after myeloablative therapy for both malignant and nonmalignant disorders.1 Recent results after unrelated CB transplantation have suggested a significant delay in immune reconstitution.2,3 CB hematopoiesis and cellular immunity are developmentally immature when compared with adult peripheral blood (APB). This immaturity may be due, in part, to defects in CB cellular lymphokine and cytokine production.4 Neonatal lymphocytes from CB have reduced T-cell5,6 and natural killer (NK) cell function7,8 and decreased type-specific antibody production.9,10 Harris et al11,12 and others have also reported that CB is composed of phenotypically and functionally immature lymphocytes that are associated with decreased allo-antigen–specific cytotoxic T lymphocytes (CTLs).13,14 We have previously shown reduced mRNA expression and protein production of specific cytokines including granulocyte-macrophage colony-stimulating factor (GM-CSF ), granulocyte colony-stimulating factor (G-CSF ), interleukin-3 (IL-3), macrophage colony-stimulating factor (M-CSF ), and IL-12 from stimulated human CB versus APB mononuclear cells (MNCs).15-18 Decreased production of tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) from human CB versus APB MNCs has also been reported.19-21 These defects in CB T-cell immune function and specific cytokine production may contribute to the delayed immune reconstitution in patients after unrelated CB transplantation.

IL-15 was recently identified as a novel cytokine that was originally cloned from a simian kidney epithelial cell line (CV-1/EBNA).22 The IL-15 gene has 8 exons and is located in the human genome on chromosome 4q31.23 Human IL-15 cDNA encodes a 162 amino acid peptide with a long leader sequence of 48 amino acids yielding a 114 amino acid mature protein with a size of 13 to 14 kD.22 IL-15 mRNA is expressed by a wide variety of tissues, including placenta, skeletal muscle, kidney, lung, heart, fibroblasts, epithelial cells, and most abundantly by activated monocytes. IL-15 shares many biologic properties with IL-2, including induction of T-cell, B-cell, and NK cell proliferation.22,24-28 Comparatively, IL-2 is predominantly expressed and produced by activated T cells.22,29 IL-15 binds to only the β and γ subunits of the IL-2 receptor complex without requiring the use of the α subunit to exert its biologic activities.22,30-32 IL-15 has been reported to enhance nonspecific NK and lymphokine-activating killer (LAK) cytotoxic activities.22,26-28 IL-15 also induces NK cell production of IFN-γ, TNF-α, and GM-CSF.28 In vitro and in vivo studies have shown IL-15 to induce a variety of antitumor effects, including induction of CTL and LAK activities.33 34 

We hypothesize that the reduced production of IL-15 from activated CB MNCs compared with APB MNCs may contribute, in part, to immaturity of CB cellular immunity and potentially the delay in immune reconstitution after unrelated CB transplantation. Furthermore, exogenous IL-15 administration may enhance CB cellular immunity and has the potential for enhancing immune reconstitution and for immunotherapy after unrelated CB transplantation. In this study, therefore, we investigated the expression and production of IL-15 and regulatory mechanisms associated with IL-15 expression in CB compared with APB MNCs. We sought to determine if IL-15 could enhance CB IFN-γ and TNF-α production and CB NK and LAK cytotoxicities compared with APB and also to determine if IL-15 would be additive or synergistic with IL-12 with regard to in vitro antitumor immunity.

Isolation of MNCs from CB and APB. Peripheral blood was obtained by venipuncture from healthy adult volunteers in accordance with the principles of the Declaration of Helsinki. Blood samples were also obtained from the umbilical cords of the placentas of normal, full-term, nonstressed infants immediately after scheduled cesarean section. The samples were collected in heparinized syringes. CB and APB MNCs were isolated from whole blood by density gradient separation on Ficoll-Hypaque gradients (density = 1.007 g/mL; Sigma Chemical Co, St Louis, MO) for 30 minutes. The MNCs at the interface were collected, washed twice, and resuspended in RPMI-1640 (GIBCO, Grand Island, NY) culture medium supplemented with 10% heat-inactivated human AB serum (Sigma). MNCs isolated by this density gradient separation were purified to greater than 98% homogeneity, and cell viability as measured by trypan blue exclusion was more than 99%. There was no difference in the MNC differential between CB and APB (CB: 82% ± 8.0% lymphocytes and 8.8% ± 4.0% monocytes; APB: 86% ± 4.0% lymphocytes and 7.2% ± 3.0% monocytes). The cells were cultured at a density of 1 × 106 cells/mL in culture medium for the following assays.

RNA isolation and Northern blotting. To determine IL-15 mRNA expression, CB and APB MNCs (60 × 106 cells) were stimulated with lipopolysaccharide (LPS; from Escherichia coli 0127:B8 at 10 μg/mL; Sigma) for up to 48 hours. Total cellular RNA was extracted from stimulated and unstimulated cells by the method of Chomczynski and Sacchi.35 Polyadenylated (A+) RNA from cytoplasmic total RNA was then purified with oligo (dT) cellulose column (mRNA purification kit from Pharmacia Biotech Inc, Piscataway, NJ) and electrophoresed on 1% agarose and 5% formaldehyde gels. The samples were heated in 40% formamide and 14% formaldehyde at 65°C for 15 minutes and then cooled before the addition of 1 μg/mL ethidium bromide. RNA was transferred to nitrocellulose and baked for 2 hours. Hybridization with an antisense probe made by transcription of a human cDNA (kindly provided by Dirk Anderson, Immunex, Seattle, WA) was performed at 63°C overnight in 50% formamide, 5× sodium chloride sodium citrate (SSC), 1× Denhardt's, 50 mmol/L sodium phosphate (pH 6.5), 0.1% sodium dodecyl sulfate (SDS), 250 μg/mL salmon sperm DNA, and 10% dextran sulfate. Blots were washed with 2× SSC at room temperature and with 0.1× SSC at 68°C for 1 hour. Blots were exposed to Kodak XAR-5 film (Eastman Kodak, Rochester, NY). The hybridization signals were quantified by densitometry of autoradiographs. Blots were then rehybridized with glyceride-3 phosphate dehydrogenase (GAPDH) probe, 775-bp Pst I/Xba I fragment from phcGAP (ATCC, Rockville, MD), as an internal standard. The levels of IL-15 mRNA were calculated by normalizing signal optical densities to those of GAPDH mRNA. Cord and adult Northern blot analyses were performed simultaneously under identical hybridization conditions and with the same amount of exposure time of the blot.

Immunoblot analysis. Plastic adherent monocytes (10 to 20 × 106 cells, >95% CD14+ monocytes) were obtained from MNC culture (5 × 106 cell/mL) after being incubated at 37°C for 1 hour according to the method of D'Andrea et al.36 Stimulated (LPS at 10 μg/mL for 12 hours) and adhered (without LPS) monocyte cultures were gently washed twice in phosphate-buffered saline (PBS). Whole cells were then lysed in PBS containing 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mmol/L EDTA, 1 mmol/L phenylmethyl sulfonyl fluoride,37 and 20 μg/mL pepstatin A for 30 minutes on ice. Lysates were spun in a microfuge at 12,000 rpm for 15 minutes. Supernatant was collected and stored at −70°C for Western blot analysis. Lysates were thawed, and quantitation of total protein from each sample was performed in duplicate, twice before loading, using the BCA protein assay (Pierce, Rockford, IL). Equal amounts (20 μg) of protein were loaded per lane, and proteins were separated by 8% SDS-polyacrylamide gel electrophoresis (SDS-PAGE). Proteins were electrophoretically transferred to nitrocellulose membranes and blocked with 5% nonfat dry milk solubilized in PBS for 1 hour at room temperature. Blots were then incubated in 10 μg equivalent of goat polyclonal antibody (AB-247-NA; R & D Systems, Minneapolis, MN) directed against human IL-15 protein in 2% milk (vol/vol, solubilized in PBS) overnight at 4°C. Incubation with 0.2% (vol/vol) horseradish peroxidase-conjugated swine antigoat IgG (BioSource International, Camarillo, CA) for 1 hour at room temperature followed. IL-15 protein bands were visualized by development with luminol/peroxide chemiluminescent substrate (Pierce) for 1 minute and exposed to x-ray film for 30 seconds. Two-dimensional densitometry of film was performed using the Bio-Image Model 50S (Millipore, Bedford, MA) automated scanning system. Immunoblot analyses were performed with three different samples.

Nuclear run-on transcription assay. Cultures of three different samples were stimulated with LPS (10 μg/mL; Sigma) for 12 hours to obtain maximal induction of IL-15. Nuclei isolation and nuclear run-on assays were performed as previously described38 and involved modification procedures described by Weber et al39 and Groudine et al.40 Nuclear run-on assays were performed with cDNA (IL-15, 0.48 kb cDNA; GAPDH, 775-bp Pst I/Xba I fragment from phcGAP) as targets. The amount of target DNA per slot was 1 μg. The hybridization mixture contained 2.5 to 5 × 107 cpm/5 mL. Run-on signal strengths were determined by densitometry of autoradiographs. The density of the bands was calculated by normalizing values with respect to the signals of internal standards (GAPDH).

mRNA half-life. Cord and adult MNCs (60 × 106 cells) were stimulated with LPS (10 μg/mL) for 12 hours before exposure to the transcriptional inhibitor, actinomycin D (10 μg/mL), as described previously.17 Cells were harvested at intervals of 0, 60, 120, 240, and 360 minutes. Cytoplasmic RNA was extracted, poly (A)+ RNA was purified, and Northern blot analysis was performed as described above for the IL-15 mRNA expression. The amount of IL-15 mRNA was normalized to the amount of GAPDH mRNA in each sample and then expressed as a percentage, setting the amount of mRNA at time 0 equal to 100%. The data were plotted against the time after addition of actinomycin D, and the half-life of each transcript was calculated based on the resultant graphs.

Induction of cytokine production and NK and LAK cytotoxicity. Recombinant simian IL-15 (specific activity on CTLL proliferation, 3.33 × 105 U/μg) was kindly provided by Dr T. Troutt (Immunex), and the recombinant human IL-12 (specific activity on phytohemagglutinin blast proliferation, 5.26 × 106 U/μg) was kindly provided by Dr S. Wolf (Genetics Institute, Cambridge, MA). MNCs at 5 × 106 cells/mL were placed into a plastic petri dish for 1 hour of incubation at 37°C to remove monocytes. Monocyte-depleted MNCs (MD MNCs) were adjusted to 1 × 106 cells/mL, seeded in 24-well flat-bottom plates containing varying concentrations of cytokines, and incubated at 37°C in a 5% CO2 humidified incubator. NK activity was measured against K562 target cells (NK-sensitive, a human erythroleukemic cell line; ATCC) after 18 hours of stimulation, and LAK activity was measured against Daudi target cells (LAK-sensitive, a human Burkitt's lymphoma; ATCC) after 72 hours of stimulation. At the end of the incubation, the effector cells were harvested, washed, and resuspended in appropriate concentrations based on the ratios of effector to target cells (E:T ratio) for the cytotoxicity assays.

Cytotoxicity assay. A standard 3-hour 51Cr-release assay41 was performed to measure the cytotoxicity. Briefly, the target cells were labeled with 100 μCi of Na2CrO4 , washed twice, and resuspended in a concentration of 5 × 104 cells/mL. One hundred microliters of each target and effector cell suspension with E:T ratios (20:1, 10:1, and 5:1) was added to a V-bottom 96-well culture plate. The mixture was centrifuged briefly and incubated at 33°C for 3 hours. At the end of the incubation, 150 μL of cell-free supernatant was collected from each well. The radioactivity was measured in a Beckman LS 1800 Liquid Scintillation Counter (Beckman, Fullerton, CA). All of the samples were run in triplicate. The percentage of lysis was calculated at each E:T ratio using the formula ([Experimental − Spontaneous Release]/[Maximum − Spontaneous Release]) × 100% and then converted to lytic units (LU; 30% target cell killing in 107 effector cells) using a computer-assisted program.42 To determine the tumoricidal spectrum of CB nonspecific cytotoxicity, two acute lymphoblastic leukemia cell lines, CCRF-CEM (T cells; ATCC) and CCRF-SB (B cells; ATCC), and two neuroblastoma cell lines, NB-100 (ATCC) and SK-N-MC (ATCC), were used as target cells.

The study of effects of IL-15 and IL-12 in combination on CB NK and LAK activities. A method previously described by DeBlaker-Hohe et al43 was used to determine if IL-15 and IL-12 in combination induces a synergistic or additive NK and LAK activity from CB MNCs. To avoid a deviation of using controls twice while comparing the cytolytic response from the combination of two cytokines with the sum of that from each single cytokine, the method was modified by subtracting the control from each single data before being applied to the calculation. Each cytotoxicity result of 17 combinations from 5 doses of IL-15 (0.1, 0.5, 1.0, 5.0, and 10 ng/mL) and 4 doses of IL-12 (0.1, 0.5, 1.0, and 10 U/mL) was compared with the sum of that from the same dose of each single cytokine using the following formula: [(Cytotoxicity in Combination − Sum of Each Cytokine Alone)/(Sum of Each Cytokine Alone)] × 100%. Based on the calculated results, the synergistic effect was arbitrarily defined as the cytolytic response from the combination of two cytokines exceeding the sum of that of each single cytokine by more than 10%, additive as 0% to 10% and nonadditive as less than 0%.

TNF-α and IFN-γ enzyme-linked immunosorbent assay (ELISA). CB and APB MNCs at a concentration of 1 × 106 cells/mL were stimulated by IL-15 (50 ng/mL) for 24, 72, and 144 hours. The supernatant was collected and the protein level was measured by ELISA (Biosource) following the manufacturer's protocol. All of the samples were run in duplicate and data were presented as the mean ± SEM. The sensitivity of the assay was 7.8 pg/mL.

Statistical analysis. Results from cytotoxicity studies and ELISA were presented as the mean ± SEM of three or more samples. Student's t-test was used for determining significant differences between two groups, and Kruskal-Wallis nonparametric ANOVA test was used for comparing multiple groups with Bartlett's test as the posttest for determining specific significant subgroups (Instat Graph Pad Software, San Diego, CA). A P value <.05 was considered significant.

Reduced IL-15 mRNA expression in stimulated cord versus adult MNCs. Northern blot analyses of CB and APB MNCs were performed simultaneously under identical conditions to compare the IL-15 mRNA expression before and after LPS stimulation. Unstimulated MNCs from both CB and APB had an undetectable expression of IL-15 mRNA. In a time course study of IL-15 mRNA expression after LPS (10 μg/mL) stimulation, IL-15 mRNA expression was induced within 6 hours upon LPS stimulation and reached a peak level at 12 hours in both CB and APB MNCs, returning to basal level after 24 hours. However, IL-15 mRNA expression in CB MNCs was only 25% ± 2.0% (mean ± SEM, n = 4, P < .05) compared with the level in APB MNCs after 12 hours of stimulation with LPS (Fig 1).

Fig. 1.

Time course of induction of IL-15 mRNA expression in cord (CB) and adult (APB) MNCs. CB and APB MNCs were isolated, cultured in RPMI, and stimulated with 10 μg/mL LPS for 0, 3, 6, 12, 24, and 48 hours. Cells were harvested and poly (A)+ RNA samples were analyzed by Northern blot analysis of IL-15 mRNA (1.5 kb). Results shown are representative of three different CB and APB RNA blot hybridizations, normalized to GAPDH signal. The lower panel bar graph represents comparative IL-15 mRNA expression from LPS-stimulated (12 hours) CB and APB MNCs. Four different CB and APB samples were analyzed after 12 hours of LPS stimulation (10 μg/mL) for IL-15 mRNA expression by Northern blot analysis. The amount of IL-15 mRNA was expressed as a percentage, setting the amount of APB mRNA equal to 100% (APB v CB, 100% v 25% ± 2.0%, P < .05, n = 4).

Fig. 1.

Time course of induction of IL-15 mRNA expression in cord (CB) and adult (APB) MNCs. CB and APB MNCs were isolated, cultured in RPMI, and stimulated with 10 μg/mL LPS for 0, 3, 6, 12, 24, and 48 hours. Cells were harvested and poly (A)+ RNA samples were analyzed by Northern blot analysis of IL-15 mRNA (1.5 kb). Results shown are representative of three different CB and APB RNA blot hybridizations, normalized to GAPDH signal. The lower panel bar graph represents comparative IL-15 mRNA expression from LPS-stimulated (12 hours) CB and APB MNCs. Four different CB and APB samples were analyzed after 12 hours of LPS stimulation (10 μg/mL) for IL-15 mRNA expression by Northern blot analysis. The amount of IL-15 mRNA was expressed as a percentage, setting the amount of APB mRNA equal to 100% (APB v CB, 100% v 25% ± 2.0%, P < .05, n = 4).

Close modal

Decreased IL-15 protein accumulation in cord versus adult MNCs. IL-15 protein production from whole cell lysates was determined by Western immunoblot analysis with a goat polyclonal anti–IL-15 antibody (Fig 2). Equal amounts of protein (20 μg) were loaded per lane after being quantitated in duplicate twice. Bands with a molecular size of 13 kD were detected from stimulated cord and adult cells. IL-15 was not detected in adhered cells without LPS. IL-15 protein production in LPS-stimulated CB monocytes (lanes 5 and 6) was only 30% ± 2.5% (mean ± SEM) of the level in APB monocytes (lanes 3 and 4; n = 3, P < .05) after 12 hours of LPS stimulation.

Fig. 2.

Detection of IL-15 in stimulated cord (CB) and adult (APB) monocytes. Immunoblot analysis of LPS (12 hours) stimulated CB and APB monocytes using anti–huIL-15 goat antibody. Equal amounts of protein (20 μg) were loaded per lane. Before loading, quantitation of total protein from each sample was performed in duplicate, twice using BCA protein assay. Lane 1, rhIL-15; lane 2, adhered monocytes; lanes 3 and 4, LPS-stimulated adult monocytes; lanes 5 and 6, LPS-stimulated cord monocytes. The lower panel bar graph represents comparative IL-15 protein production from three different LPS-stimulated (12 hours) CB and APB monocytes. The amount of IL-15 protein was expressed as a percentage, setting the amount of APB level equal to 100% (APB v CB, 100% v 30% ± 2.5%, P < .05, n = 3).

Fig. 2.

Detection of IL-15 in stimulated cord (CB) and adult (APB) monocytes. Immunoblot analysis of LPS (12 hours) stimulated CB and APB monocytes using anti–huIL-15 goat antibody. Equal amounts of protein (20 μg) were loaded per lane. Before loading, quantitation of total protein from each sample was performed in duplicate, twice using BCA protein assay. Lane 1, rhIL-15; lane 2, adhered monocytes; lanes 3 and 4, LPS-stimulated adult monocytes; lanes 5 and 6, LPS-stimulated cord monocytes. The lower panel bar graph represents comparative IL-15 protein production from three different LPS-stimulated (12 hours) CB and APB monocytes. The amount of IL-15 protein was expressed as a percentage, setting the amount of APB level equal to 100% (APB v CB, 100% v 30% ± 2.5%, P < .05, n = 3).

Close modal

Comparable transcriptional rate of the IL-15 gene in cord versus adult MNCs. Nuclear run-on transcriptional analysis was performed to determine if the low amount of IL-15 mRNA in stimulated CB MNCs was due to a decreased transcription rate of the IL-15 gene. Nuclear run-on transcripts from nuclei isolated from CB MNCs and stimulated with LPS for 12 hours were compared with run-on transcripts isolated from similarly treated APB MNCs. As shown in Fig 3, unstimulated CB and APB MNCs showed low basal level signals of IL-15 transcript (optical density [OD], < 0.1), which was approximately the same in both cord and adult. After stimulation with LPS (12 hours), the transcriptional rate of the IL-15 gene was significantly increased in both CB and APB MNCs (cord, 20- ± 2.5-fold; adult, 22- ± 1.7-fold; n = 3, P < .05). However, there was no appreciable difference between activated CB and APB MNC in the degree of transcriptional activation (P = not significant [NS]).

Fig. 3.

Nuclear run-on analyses of IL-15 transcription in cord (CB) versus adult (APB) MNCs stimulated with LPS (10 μg/mL for 12 hours). Equivalent amounts of radioactive labeled RNA were hybridized to filters containing the indicated target DNA. Results shown are representative of three different experiments. US, unstimulated; S, stimulated; pUC 18, control vector; GAPDH, 775-bp Pst I/Xba I fragment from phc GAP; IL-15, 480-bp cDNA (CB 20- ± 2.5-fold v APB 22- ± 1.7-fold, P = NS, n = 3).

Fig. 3.

Nuclear run-on analyses of IL-15 transcription in cord (CB) versus adult (APB) MNCs stimulated with LPS (10 μg/mL for 12 hours). Equivalent amounts of radioactive labeled RNA were hybridized to filters containing the indicated target DNA. Results shown are representative of three different experiments. US, unstimulated; S, stimulated; pUC 18, control vector; GAPDH, 775-bp Pst I/Xba I fragment from phc GAP; IL-15, 480-bp cDNA (CB 20- ± 2.5-fold v APB 22- ± 1.7-fold, P = NS, n = 3).

Close modal

Decreased IL-15 mRNA half-life in cord versus adult. Because the transcriptional rate of the IL-15 gene was virtually the same for both CB and APB MNCs, the stability of IL-15 mRNA was compared in stimulated CB and APB MNCs by blocking mRNA synthesis with actinomycin D to determine whether the differential regulation was occurring at the posttranscriptional level. CB and APB MNCs were stimulated with LPS (10 μg/mL) for 12 hours before actinomycin D (10 μg/mL) was added for various time periods (0 to 360 minutes.). Northern blots of poly (A)+ RNA were hybridized with an antisense riboprobe made by transcription of human cDNA. The levels of IL-15 mRNA progressively decreased during actinomycin D exposure in both CB and APB MNCs. Transcripts were quantitated by densitometric scanning of the autoradiographs. As shown in Fig 4, the measured mRNA half-life of IL-15 from stimulated CB MNCs was approximately twofold lower than that from stimulated APB MNCs (t1/2 : 101 ± 5.8 minutes v 210 ± 8.2 minutes, CB v APB, mean ± SEM, n = 3, P < .05).

Fig. 4.

The half-life of IL-15 mRNA in cord (CB) (A) versus adult (APB) (B) MNCs. Actinomycin D (10 μg/mL) was added for the indicated times to cells from cord (A) and adult (B), stimulated with LPS (10 μg/mL) for 12 hours. Poly (A)+ RNA was analyzed by Northern blotting for the presence of IL-15 transcript (1.5 kb). The data were plotted against the time after the addition of actinomycin D. Results shown are representative of three different experiments (t1/2 : 101 ± 5.8 minutes v 210 ± 8.2 minutes, CB v APB, P < .05, n = 3).

Fig. 4.

The half-life of IL-15 mRNA in cord (CB) (A) versus adult (APB) (B) MNCs. Actinomycin D (10 μg/mL) was added for the indicated times to cells from cord (A) and adult (B), stimulated with LPS (10 μg/mL) for 12 hours. Poly (A)+ RNA was analyzed by Northern blotting for the presence of IL-15 transcript (1.5 kb). The data were plotted against the time after the addition of actinomycin D. Results shown are representative of three different experiments (t1/2 : 101 ± 5.8 minutes v 210 ± 8.2 minutes, CB v APB, P < .05, n = 3).

Close modal

Enhanced CB and APB NK and LAK cytotoxic activities after IL-15 stimulation. The baseline NK and LAK cytotoxicities were determined after 18 and 72 hours of incubation in the absence of cytokines. The NK activity of CB against K562 was significantly lower than that of APB MD MNCs (CB v APB: 60 ± 9 v 115 ± 18 LU, P < .05, n = 10; Fig 5). However, the LAK activity of CB was similar to APB (CB v APB: LAK, 43 ± 10 v 44 ± 19 LU, P = NS, n = 10; Fig 6). After incubation of MD MNCs with IL-15 (10 ng/mL), both CB and APB NK activities (18 hours) were significantly increased over control (control v IL-15 at 10 ng/mL: CB, 45 ± 9 v 533 ± 72 LU, P < .01, n = 10; APB, 115 ± 18 v 537 ± 68 LU, P < .05, n = 6). CB NK activity reached a comparable level of APB (IL-15 at 10 ng/mL: CB v APB, 533 ± 72 v 539 ± 68 LU, P = NS). IL-15 (10 ng/mL) also induced a significant increase of both CB and APB LAK activities against Daudi target cells (control v IL-15 at 10 ng/mL: CB, 43 ± 10 v 843 ± 64 LU, P < .01, n = 12; APB, 44 ± 19 v 420 ± 107 LU, P < .01, n = 6; Fig 6). However, CB MD MNCs was more responsive to IL-15 stimulation than APB for induction of LAK activity (CB v APB: IL-15 at 1.0 ng/mL, 262 ± 62 v 144 ± 91, P < .05; IL-15 at 10 ng/mL 843 ± 64 v 420 ± 107 LU, P < .01; Fig 6).

Fig. 5.

IL-15 induction of CB and APB MNC NK cytotoxicity against K562 line. *CB v APB, 45 ± 9 v 115 ± 18 LU, P < .05, n = 10; (▨) CB; () APB.

Fig. 5.

IL-15 induction of CB and APB MNC NK cytotoxicity against K562 line. *CB v APB, 45 ± 9 v 115 ± 18 LU, P < .05, n = 10; (▨) CB; () APB.

Close modal
Fig. 6.

IL-15 induction of CB and APB MNC LAK activity against Daudi cell line. CB v APB: *IL-15 at 1 ng/mL, 258 ± 55 v 132 ± 51, P < .05; **IL-15 at 10 ng/mL, 843 ± 64 v 420 ± 107 LU, P < .01. (▧) CB; () APB.

Fig. 6.

IL-15 induction of CB and APB MNC LAK activity against Daudi cell line. CB v APB: *IL-15 at 1 ng/mL, 258 ± 55 v 132 ± 51, P < .05; **IL-15 at 10 ng/mL, 843 ± 64 v 420 ± 107 LU, P < .01. (▧) CB; () APB.

Close modal

IL-15 induced nonspecific tumoricidal NK and LAK activities. Four cell lines, CCRF-CEM, CCRF-SB, NB-100, and SK-N-MC, were used to examine in vitro antitumor activity of CB NK and LAK cells after IL-15 stimulation. IL-15 (10 ng/mL) stimulation resulted in enhanced CB cytotoxicity against all of these tumor cell lines. As shown in Fig 7A, three cell lines, CCRF-CEM, SK-N-MC, and NB-100, became more sensitive to CB NK cytotoxicity induced by IL-15 (control v IL-15 stimulated: CCRF-CEM, 50 ± 34 v 229 ± 34 LU, P < .05; SK-N-MC, 18 ± 17 v 269 ± 91 LU, P < .01; NB-100, 22 ± 4 v 208 ± 25 LU, P < .01, n = 4). IL-15–induced CB LAK cytotoxicity produced an enhanced lytic response against all four tumor cell lines over controls (control v IL-15 stimulated: CCRF-CEM, 55 ± 39 v 318 ± 3 LU, P < .01; CCRF-SB, 18 ± 12 v 436 ± 114 LU, P < .05; SK-N-MC, 50 ± 25 v 358 ± 13 LU, P < .01; NB-100, 33 ± 31 v 330 ± 40 LU, P < .01, n = 3; Fig 7B).

Fig. 7.

(A and B) IL-15 (10 ng/mL) induction of CB NK and LAK cytotoxicity against several tumor cell lines. Acute lymphoblastic leukemia cell lines: CCRF-CEM (T-lymphoblastoid cells) and CCRF-SB (B-lymphoblastoid cells); neuroblastoma cell lines: SK-N-MC and NB-100. (A) The tumoricidal spectrum of CB NK activity: IL-15 (10 ng/mL) v unstimulated: CCRF-CEM*, 229 ± 75 v 50 ± 34 LU; SK-N-MC**, 269 ± 91 v 18 ± 17 LU; NB-100**, 208 ± 25 v 22 ± 4 LU; n = 4. (▨) Medium; () IL-15 (10 ng/mL). (B) The tumoricidal spectrum of CB LAK activity: IL-15 (10 ng/mL) v unstimulated: CCRF-CEM**, 318 ± 3 v 55 ± 39 LU; CCRF-SB*, 436 ± 114 v 18 ± 12 LU; SK-N-MC**, 358 ± 13 v 50 ± 25 LU; NB-100**, 330 ± 40 v 33 ± 31 LU; n = 3. (▨) Medium; (▩) IL-15 (10 ng/mL). *P < .05; **P < .01.

Fig. 7.

(A and B) IL-15 (10 ng/mL) induction of CB NK and LAK cytotoxicity against several tumor cell lines. Acute lymphoblastic leukemia cell lines: CCRF-CEM (T-lymphoblastoid cells) and CCRF-SB (B-lymphoblastoid cells); neuroblastoma cell lines: SK-N-MC and NB-100. (A) The tumoricidal spectrum of CB NK activity: IL-15 (10 ng/mL) v unstimulated: CCRF-CEM*, 229 ± 75 v 50 ± 34 LU; SK-N-MC**, 269 ± 91 v 18 ± 17 LU; NB-100**, 208 ± 25 v 22 ± 4 LU; n = 4. (▨) Medium; () IL-15 (10 ng/mL). (B) The tumoricidal spectrum of CB LAK activity: IL-15 (10 ng/mL) v unstimulated: CCRF-CEM**, 318 ± 3 v 55 ± 39 LU; CCRF-SB*, 436 ± 114 v 18 ± 12 LU; SK-N-MC**, 358 ± 13 v 50 ± 25 LU; NB-100**, 330 ± 40 v 33 ± 31 LU; n = 3. (▨) Medium; (▩) IL-15 (10 ng/mL). *P < .05; **P < .01.

Close modal

Additive and synergistic effects of low doses of IL-15 and IL-12 on CB NK and LAK cytotoxicity. The effects of the combination of IL-15 and IL-12 on NK and LAK cytotoxicity was examined by comparing the cytolytic response from two cytokines in combination with the sum of that from each single cytokine. The results in Table 1 show that using lower doses of IL-15 (0.1, 0.5, and 1.0 ng/mL) and IL-12 (0.1, 0.5, and 1.0 U/mL) concomitantly generated either synergistic or additive effects on CB NK cytotoxicity against K562 targets. The synergy from these lower dose combinations induced a comparable NK cytotoxicity compared with single higher doses of each cytokine individually. Specifically, IL-15 (1.0 ng/mL) and IL-12 (0.5 U/mL) induced a lytic response comparable to that induced by 10 ng/mL of IL-15 alone or 10 U/mL of IL-12 alone (532 ± 64 v 473 ± 70 LU, P = NS, and v 410 ± 48 LU, P = NS). Conversely, higher dose combinations of IL-12 and IL-15 induced only nonadditive effects on CB NK cytotoxicity. Table 2 summarizes the combined effects of IL-15 and IL-12 on CB LAK cytotoxicity against Daudi targets. The synergistic effect of IL-12 and IL-15 is seen at two combinations of lower doses of IL-15 and IL-12 (0.1 or 0.5 ng/mL of IL-15 + 0.1 U/mL of IL-12) and the lytic response from the latter combination is comparable to that of a single higher dose of IL-12 (10 U/mL), but much lower than that of IL-15 (10 ng/mL) (IL-15 + IL-12 v IL-12 alone v IL-15 alone: 512 ± 48 LU v 501 ± 54 LU, P = NS; and v 823 ± 68 LU, P < .01). The higher dose combinations of IL-15 and IL-12 induced a suppressive effect on LAK cytotoxicity when compared with the single higher dose of IL-12 and IL-15 (10 ng/mL IL-15 + 10 U/mL IL-12 v 10 ng/mL IL-15 alone v 10 U/mL IL-12 alone, 259 ± 77 v 823 ± 68 LU, P < .01; 259 ± 77 v 501 ± 54, P < .05, n = 10).

IL-15 enhancement of IFN-γ and TNF-α production from CB MNCs. IL-15 (50 ng/mL) stimulation induced a significant increase of IFN-γ protein production in CB MNCs (IL-15 stimulated v control: day 1, 112 ± 13 v 3.3 ± 1.2 pg/mL, P < .01; day 3, 480 ± 75 v 8 ± 4 pg/mL, P < .01; day 6, 670 ± 20 v 135 ± 52 pg/mL, P < .01, n = 3) However, the IFN-γ levels in IL-15–stimulated CB were still significantly lower than stimulated APB (IL-15–stimulated CB v IL-15–stimulated APB: day 1, 112 ± 13 v 567 ± 44 pg/mL, P < .01; day 3, 480 ± 75 v 866 ± 30 pg/mL, P < .05; day 6, 670 ± 20 v 830 ± 20 pg/mL, P < .05, n = 3; Fig 8A). Similarly, IL-15 (50 ng/mL) stimulation also significantly increased TNF-α protein production in CB MNCs (IL-15 stimulated v control: day 1, 308 ± 81 v 50 ± 26 pg/mL, P < .01; day 3, 423 ± 76 v 70 ± 30 pg/mL, P < .01; day 6, 358 ± 44 v 128 ± 51 pg/mL, P < .05, n = 3). The TNF-α production in CB MNCs was still significantly lower than APB (IL-15–stimulated CB v stimulated APB: day 1, 308 ± 46 v 676 ± 88 pg/mL, P < .01; day 3, 423 ± 76 v 983 ± 179 pg/mL, P < .05; day 6, 358 ± 44 v 1,125 ± 108, P < .01, n = 3; Fig 8B).

Fig. 8.

(A and B) IL-15 (50 ng/mL) induction of IFN-γ and TNF-α production in CB MNCs. (A) Time course of IFN-γ production in CB and APB MNCs; (▨) CB-control, (▨) CB–IL-15, (▧) APB-control, (▧) APB–IL-15. (B) Time course of TNF-α production in CB and APB MNCs; (▩) CB-control, () CB–IL-15, ()]) APB-control, ()]) APB–IL-15.

Fig. 8.

(A and B) IL-15 (50 ng/mL) induction of IFN-γ and TNF-α production in CB MNCs. (A) Time course of IFN-γ production in CB and APB MNCs; (▨) CB-control, (▨) CB–IL-15, (▧) APB-control, (▧) APB–IL-15. (B) Time course of TNF-α production in CB and APB MNCs; (▩) CB-control, () CB–IL-15, ()]) APB-control, ()]) APB–IL-15.

Close modal

Numerous studies have demonstrated the success of hematopoietic growth factors to enhance hematologic reconstitution after stem cell transplantation.44-51 Vowels et al52 reported that the use of GM-CSF post-CB transplantation resulted in rapid engraftment and mild graft-versus-host disease. However, the delay in immune reconstitution post-CB transplantation may be due to the defects in CB cellular immunity and cytokine production.2-4 We have recently reported that the reduced expression and production of IL-12 from activated CB may contribute to the immaturity in CB cellular immunity.18 The number and functionality of donor-derived lymphocytes in patients after CB transplantation remains to be determined. The use of exogenous cytokines post-CB transplantation that enhance CB immune function may compensate for the immaturity of CB cellular immunity and enhance immune reconstitution and CB tumor immunity post-CB transplantation.

Although there are many similar biologic activities between IL-2 and IL-15, the regulation of IL-15 expression differs markedly from that of IL-2. IL-15 mRNA is expressed in a variety of tissues, including placenta, skeletal muscle, kidney, and activated monocytes/macrophages but not normal T cells.22 Enhancement of IL-15 protein production from monocytes occurs in response to a wide variety of agonists including LPS, IFN-γ, Bacillus Calmette-Guerlin (BCG), Mycobacterium tuberculosis, Toxoplasma gondii, or Salmonella cholersesuis.53-55 During states of increased demand (stimulation), mononuclear phagocytes contribute significantly to the production of IL-15 in both CB and APB. However, our present results suggest that CB MNCs express and produce less IL-15 in response to bacterial stimulation (Figs 1 and 2). The decreased IL-15 mRNA expression in CB versus APB MNCs is not secondary to alteration in IL-15 gene transcription (Fig 3). In comparison, alterations in posttranscriptional stability appear to account, in part, for the decrease in IL-15 mRNA expression in CB versus APB MNCs (Fig 4).

Control of mRNA stability is not well understood, but the process is thought to involve various factors interacting with specific mRNA sequences.56,57 The adenosine + uridine (AU)-rich or AUUUA-repeat elements (ARE) in the 3′ untranslated regions (UTR) of many cytokine and protooncogene transcripts are known to be the targets of a pathway for selective processing and mRNA degradation.58-62 Several AUUUA repeats of the 3′ UTR of IL-15 sequence may contribute to the instability of IL-15 mRNA.

We previously reported the complex posttranscriptional regulatory mechanisms associated with several cytokines. The reduced accumulation of M-CSF, GM-CSF, and IL-12 mRNA in CB MNCs is associated with a reduction in mRNA half-life compared with APB MNCs, whereas the rate of gene transcription remains comparable in CB and APB MNCs.15,17,18 The 3′ UTR of these cytokines have multiple copies of AUUUA-elements.63,64 A reduced mRNA half-life and comparable transcription rates for GM-CSF, M-CSF, and IL-12 in CB versus APB MNCs indicate that these ARE-containing transcripts may also be less stable in CB MNCs. Translational inhibition by cycloheximide after stimulation of cells causes a superinduction of GM-CSF, as well as M-CSF mRNA, which is approximately 2.5-fold greater in CB versus APB MNCs.17,65 Increased transcript stabilization in stimulated CB MNCs after CHX treatment suggests that, before translational inhibition, higher levels of a translational-dependent nuclease may be present in CB MNCs. An ARE-directed endonuclease66 and several ARE-binding factors61,62,67-71 have been identified. One of these is a 37-kD protein designated AUF1. We have recently reported that the decreased GM-CSF mRNA stability in CB versus APB MNCs was inversely correlated with AUUUA-element binding activity and with the levels of AUF1 binding factor.65 It seems likely that various protein factors interacting with specific mRNA sequences exist in vivo and are involved in the regulation of AU-rich mRNA decay. Any alteration in the expression and/or biologic activities of these various protein factors in stimulated CB MNCs could contribute to the reduction of IL-15 mRNA expression. Further studies are required to test these possibilities.

The immaturity of CB immunity, which is associated with decreased production of IL-2, IL-12, IL-15, IFN-γ, and TNF-α, may contribute to diminished CB NK, LAK, and CTL cytotoxicities. Seki et al7 and others have reported that NK cytotoxicity is decreased in CB compared with APB. IL-2 can enhance the NK cytotoxicity of CB MNCs to the level of APB MNC activity.11 We have recently18 reported that IL-12 can enhance CB NK cytotoxicity up to levels of APB MNC activity. IL-15 not only enhances T-cell function, but also enhances cytolytic function of both CD56dim NK and CD8+ T cells.22,28,32 Carson et al28 reported that activation of CD56dim NK cells by IL-15 was similar to that of IL-2. However, the IL-15–enhanced NK cytotoxic activity is completely IL-2 independent. Our present studies suggested that IL-15 also enhanced CB NK and LAK activities up to the adult level. CB LAK activity appeared to be more sensitive to exogenous IL-15 compared with APB (Fig 5). Tumoricidal studies showed that IL-15 induced significant CB NK and LAK activities against a broad range of neuroblastoma, leukemia, and lymphoma cell lines (Fig 7).

Although IL-2 has been shown to have therapeutic benefits for some cancer patients,72 the substantial toxicities associated with high doses of IL-2 have limited its use clinically.73 IL-12 has also experienced dose-limiting toxicities.74 Recently, IL-15 has been shown to mimic the antitumor activities of IL-2 with potentially less toxicity in an in vivo animal model.33 Further studies are required to evaluate this aspect. Combinations of lower doses of IL-15 and IL-12 might have the potential of augmenting in vivo antitumor immune function and minimizing toxicity. DeBlaker-Hoke et al43 showed a synergistic effect on inducing APB NK and LAK activities by the combination of lower doses of IL-2 and IL-12. Carson et al28 suggested that the combination of IL-12 and IL-15 had a synergistic effect on augmenting APB NK cytolytic activity and IFN-γ production. In the present study, we demonstrated that low-dose combinations of IL-12 (0.1 U/mL) and IL-15 (0.1 to 1.0 ng/mL) induced a synergistic or additive effect on CB NK cytotoxicity, except for the combination of IL-12 (1.0 U/mL) and IL-15 (1.0 ng/mL). The synergistic NK activity reached the same levels as a single high dose (IL-12, 10 U/mL; IL-15, 10 ng/mL) of either individual cytokine. Although no synergistic or additive effects from high-dose combinations of IL-12 (5 to 10 U/mL) and IL-15 (5 to 10 ng/mL) on CB NK activity are seen, the cytotoxicity levels were still higher than that induced by the single dose of individual cytokine (Table 1). Similarly, low-dose combinations of IL-12 (0.1 U/mL) and IL-15 (0.1 to 0.5 ng/mL) had a synergistic effect on CB LAK activity that is comparable to the level as a single high dose of IL-12 (10 U/mL), but lower than a single high dose of IL-15 (10 to 100 ng/mL). However, the high-dose combination of IL-12 (1.0 to 10 U/mL) and IL-15 (10 to 100 ng/mL) had a suppressive effect on CB LAK activity (Table 2). This suppression may be due to NK cell apoptosis in which decreased numbers of killer cells resulted in a low level of LAK activity. This observation is consistent with the report by Ross and Caligiuri75 in which costimulation of IL-12 and IL-15 or IL-12 and IL-2 induced NK cell proliferation and IFN-γ production initially, followed by NK cell apoptosis and a decline in IFN-γ production.

The IFN-γ and TNF-α production in IL-15–stimulated CB MNCs was significantly induced and increased up to the unstimulated APB level, but far lower than the IL-15 stimulated APB level (Fig 8). This result is consistent with our earlier observation on IFN-γ production in IL-12–stimulated CB and APB MNCs.18 This partial compensation after IL-15 stimulation suggests that the decreased production of IFN-γ and TNF-α by CB MNCs may be due to at least two factors: defective CB IFN-γ and TNF-α production and defective CB IFN-γ– and TNF-α–inducing cytokines, such as IL-15, IL-12, and IL-2. Interestingly, the effect of IL-15 on CB NK activity and IFN-γ production showed that IL-15 alone is capable of inducing CB NK activity up to the APB level; however, IL-15 alone cannot compensate for IFN-γ production up to the stimulated APB level (Figs 5 and 8). This disparity indicates the important role of intrinsically deficient cytokine production such as IL-12, IL-15, and IFN-γ in the pathogenes of the immaturity of CB cellular immunity. Further studies are required to verify these observations.

In summary, the present study showed that IL-15 mRNA and protein production is decreased in activated CB compared with APB MNCs. This discrepancy in IL-15 production is secondary, at least in part, to altered posttranscriptional regulation. The reduced production of IL-15 from activated CB MNCs might contribute to the immaturity in CB cellular immunity. However, exogenous IL-15 stimulation may compensate for the immaturity in CB immunity by enhancing NK and LAK activities and by inducing IFN-γ and TNF-α production. The additional synergistic effects of lower doses of IL-15 in combination with IL-12 suggests the potential benefit of the combination of each cytokine to increase CB antitumor immunity and potentially decrease toxicity compared with higher doses of either of the cytokines alone. Further studies are needed to define the clinical implications of these findings and the potential use of IL-15 to enhance CB cellular immunity and/or accelerate immune reconstitution after CB transplantation.

The authors thank Sally Anderson, Renee Dulak, and Linda Rahl for their editorial assistance in the preparation of this manuscript.

J.X.Q., S.m.L., and Y.S. contributed equally to the manuscript.

Supported by grants from the Pediatric Cancer Research Foundation and the Walden W. and Jean Young Shaw Foundation.

Presented in part at the American Society of Hematology, December 1996, Orlando, FL.

Address reprint requests to Mitchell S. Cairo, MD, Director, Hematology/Oncology Research and Blood and Marrow Transplantation, Children's Hospital of Orange County, 455 S Main St, Orange, CA 92868.

1
Wagner
JE
Rosenthal
J
Sweetman
R
Shu
XO
Davies
SM
Ramsay
NKC
McGlave
PB
Sender
L
Cairo
MS
Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: Analysis of engraftment and acute graft-versus-host disease.
Blood
88
1996
795
2
Kurtzberg
J
Laughlin
M
Graham
ML
Smith
C
Olson
JF
Halperin
EC
Ciocci
G
Carrier
C
Stevens
C
Rubinstein
P
Placental blood as a source of hemtopoietic stem cells for transplantation into unrelated recipients.
N Eng J Med
335
1996
157
3
Rosenthal J, Sweetman R, Sender S, Murphy L, Slone V, Cairo MS: Immunological reconstitution after unrelated cord blood (CB) transplantation. Pediatr Res 39:161A, 1996 (abstr)
4
Cairo
M
Therapeutic implications of dysregulated colony-stimulating factor expression in neonates.
Blood
82
1993
2269
5
Monoz
AI
Limbert
D
Skin reactivity to Candida and streptokinase-streptodornase antigens in normal pediatric subjects: Influence of age and acute illness.
J Pediatr
88
1977
975
6
Franz
ML
Carella
JA
Galant
SP
Cutaneous delayed hypersensitivity in a healthy pediatric population: Diagnostic value of diphtheria-tetanus toxoids.
J Pediatr
88
1976
978
7
Seki
H
Ueno
Y
Taga
K
Matsuda
A
Miyawaki
T
Taniguchi
N
Mode of in vitro augmentation of natural killer cell activity by recombinant human interleukin 2: A comparative study of LEU-11+ and LEU-11− cell populations in cord blood and adult peripheral blood.
J Immunol
135
1985
2351
8
Baley
JE
Schacter
BZ
Mechanisms of diminished natural killer cell activity in pregnant women and neonates.
J Immunol
134
1985
3042
9
Miyawaki
T
Moriya
N
Nagaoki
T
Taniguchi
N
Maturation of B-cell differentiation ability and T-cell regulatory function in infancy and childhood.
Immunol Rev
57
1981
61
10
Palacios
R
Andersson
U
Autologous mixed lymphocyte reaction in human cord blood lymphocytes: Decreased generation of helper and cytotoxic T-cell functions and increased proliferative response and induction of suppressor T cells.
Cell Immunol
66
1982
88
11
Harris
D
Schumacher
M
Locascio
J
Besencon
F
Olson
G
DeLuca
D
Shenker
L
Bard
J
Boyse
E
Phenotypic and functional immaturity of human umbilical cord blood T lymphocytes.
Proc Natl Acad Sci USA
89
1992
10006
12
Harris
DT
LoCascio
J
Besencon
FJ
Analysis of the alloreactive capacity of human umbilical cord blood: Implications for graft-versus-host disease.
Bone Marrow Transplant
14
1994
545
13
Risdon
G
Gaddy
J
Stehman
FB
Broxmeyer
HE
Proliferative and cytotoxic responses of human cord blood T lymphocytes following allogeneic stimulation.
Cell Immunol
154
1994
14
14
Risdon
G
Gaddy
J
Horie
M
Broxmeyer
HE
Alloantigen priming induces a state of unresponsiveness in human umbilical cord blood T cells.
Proc Natl Acad Sci USA
92
1995
2413
15
Cairo
M
Suen
Y
Knoppel
E
van de Ven
C
Nguyen
A
Sender
L
Decreased stimulated GM-CSF production and GM-CSF gene expression but normal numbers of GM-CSF receptors in human term newborns compared to adults.
Pediatr Res
30
1991
362
16
Cairo
M
Suen
Y
Knoppel
E
Dana
R
Park
L
Clark
S
van de Ven
C
Sender
L
Decreased G-CSF and IL-3 production and gene expression from mononuclear cells of newborn infants.
Pediatr Res
31
1992
574
17
Suen
Y
Lee
S
Schreurs
J
Knoppel
E
Cairo
MS
Decreased macrophage colony-stimulating factor mRNA expression from activated cord versus adult mononuclear cells: Altered post transcriptional stability.
Blood
84
1994
4269
18
Lee
SM
Suen
Y
Chang
L
Bruner
V
Qian
J
Indes
J
Knoppel
E
van de Ven
C
Cairo
MS
Decreased interleukin-12 (IL-12) from activated cord versus adult peripheral blood mononuclear cells and upregulation of interferon-γ, natural killer, and lymphokine-activated killer activity by IL-12 in cord blood mononuclear cells.
Blood
88
1996
945
19
English
K
Burchett
S
English
J
Ammann
A
Wara
D
Wilson
C
Production of lymphotoxin and tumor necrosis factor by human neonatal mononuclear cells.
Pediatr Res
24
1988
717
20
Weatherstone
K
Rich
E
Tumor necrosis factor/cachectin and interleukin-1 secretion by cord blood monocytes from premature and term neonates.
Pediatr Res
25
1989
342
21
Wilson
C
Westall
J
Johnston
L
Lewis
D
Dower
S
Alpert
A
Decreased production of interferon-gamma by human neonatal cells.
J Clin Invest
77
1986
860
22
Grabstein
KH
Eisenman
J
Shanebeck
K
Rauch
C
Srinivasan
S
Fung
V
Beers
C
Richardson
J
Schoenborn
MA
Ahdieh
M
Johnson
L
Alderson
M
Watson
JD
Anderson
DM
Giri
JG
Cloning of a T cell growth factor that interacts with the β chain of the interleukin-2 receptor.
Science
264
1994
965
23
Anderson
DM
Johnson
L
Glaccum
M
Copelan
NG
Gilbert
DJ
Jenkins
NA
Valentine
V
Kirstein
MN
Shapiro
DN
Morriss
SW
Grabstein
K
Cosman
D
Chromosomal assignment and genomic structure of IL-15.
Genomics
25
1995
701
24
Armitage
RJ
Macduff
BM
Eisenman
J
Paxton
R
Grabstein
KH
IL-15 has stimulatory activity for the induction of B cell proliferation and differentiation.
J Immunol
154
1995
483
25
Mrozek
E
Anderson
P
Caligiuri
MA
Role of interleukin-15 in the development of human CD56+ natural killer cells from CD34+ hematopoietic progentior cells.
Blood
87
1996
2632
26
Burton
JD
Bamford
RN
Peters
C
Grant
AJ
Kurys
G
Goldman
CK
Brennan
J
Roessler
E
Waldmann
TA
A lymphokine, provisionally designated interleukin T and produced by a human adult T-cell leukemia line, stimulates T-cell proliferation and the induction of lymphokine-activated killer cells.
Proc Natl Acad Sci USA
91
1994
4935
27
Bamford
RN
Grant
AJ
Burton
JC
Peters
C
Kurys
G
Goldman
CK
Brennan
J
Roessler
E
Waldmann
TA
The interleukin (IL) 2 receptor β chain is shared by IL-2 and a cytokine, provisionally designated IL-T, that stimulates T-cell proliferation and the induction of lymphokine-activated killer cells.
Proc Natl Acad Sci USA
91
1994
4940
28
Carson
WE
Giri
JG
Lindemann
MJ
Linett
ML
Ahdieh
M
Paxton
R
Anderson
D
Eisenmann
J
Grabstein
K
Caligiuri
MA
Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor.
J Exp Med
180
1994
1395
29
Bamford
RN
Battiata
AP
Burton
JD
Sharma
H
Waldmann
TA
Interleukin (IL) 15/IL-T production by the adult T-cell leukemia cell line HuT-102 is associated with a human T-cell lymphotrophic virus type I R region/IL-15 fusion message that lacks many upstream AUGs that normally attenuate IL-15 mRNA translation.
Proc Natl Acad Sci USA
93
1996
2897
30
de Jong
J
Farner
NL
Widner
MB
Giri
JG
Sondel
PM
Interaction of IL-15 with the shared IL-2 receptor β and γc subunits.
J Immunol
156
1996
1339
31
Matthews
DJ
Clark
PA
Herbert
J
Morgan
G
Armitage
RJ
Kinnon
C
Minty
A
Grabstein
KH
Caput
D
Ferrara
P
Callard
R
Function of the interleukin-2 (IL-2) receptor γ-chain in biologic responses of x-linked severe combined immunodeficient B cells to IL-2, IL-4, IL-13, and IL-15.
Blood
85
1995
38
32
Giri
JG
Ahdieh
M
Eisenman
J
Shanebeck
K
Grabstein
K
Kumaki
S
Namen
A
Park
LS
Cosman
D
Anderson
D
Utilization of the β and γ chains of the IL-2 receptor by the novel cytoine IL-15.
J EMBO
13
1994
2822
33
Munger
W
DeJoy
SQ
Jeyaseelan
R Sr
Torley
LW
Grabstein
KH
Eisenmann
J
Paxton
R
Cox
T
Wick
MM
Kerwar
SS
Studies evaluating the antitumor activity and toxicity of interleukin-15, a new T cell growth factor: Comparison with interleukin-2.
Cell Immunol
165
1995
289
34
Gamero
AM
Ussery
D
Reintgen
DS
Puleo
CA
Djeu
JY
Interleukin 15 induction of lymphokine-activated killer cell function against autologous tumor cells in melanoma patient lymphocytes by a CD18-dependent, perforin-related mechanism.
Cancer Res
55
1995
4988
35
Chomczynski
P
Sacchi
N
Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction.
Anal Biochem
162
1987
156
36
D'Andrea
A
Rengaraju
M
Valiante
NM
Chehimi
J
Kubin
M
Aste
M
Chan
SH
Kobayashi
M
Young
D
Nickbarg
E
Chizzonite
R
Wolf
SF
Trinchieri
G
Production of natural killer cell stimulatory factor (interleukin 12) by peripheral blood mononuclear cells.
J Exp Med
176
1992
1387
37
Biro
S
Fu
Y-M
Yu
Z-X
Epstein
S
Inhibitory effects of antisense oligodeoxynucleotides targeting c-myc mRNA on smooth muscle cell proliferation and migration.
Proc Natl Acad Sci USA
90
1993
654
38
Lee
S
Knoppel
E
van de Ven
C
Cairo
M
Transcriptional rates of granulocyte-macrophage colony-stimulating factor, granulocyte colony-stimulating factor, interleukin-3 and macrophage colony-stimulating factor genes in activated cord vs. adult mononuclear cells: Alteration in cytokine expression may be secondary to post-transcriptional instability.
Pediatr Res
34
1993
560
39
Weber
B
Horiguchi
J
Luebbers
R
Sherman
M
Kufe
D
Posttranscriptional stabilization of c-fms mRNA by a labile protein during human monocytic differentiation.
Mol Cell Biol
9
1989
769
40
Groudine
M
Peretz
M
Weintraub
H
Transcriptional regulation of hemoglobin switching in chicken embryos.
Mol Cell Biol
1
1981
281
41
Martz E: The 51CR-release assay for CTL-mediated target cell lysis, in Sitkovsky M, Henkart P (eds): Cytotoxic Cells, vol 43. Boston, MA, Birkhauser Boston, 1993, p 457
42
Pross
HF
Baines
MG
Rubin
P
Shragge
P
Patterson
MS
Spontaneous human lymphocyte-mediated cytotoxicity against tumor target cells. IX. The quantitation of natural killer cell activity.
J Clin Immunol
1
1981
51
43
DeBlaker-Hohe
DF
Yamauchi
A
Yu
C-R
Horvath-Arcidiacono
JA
Bloom
ET
IL-12 synergized with IL-2 to induce lymphokine-activated cytotoxicity and perforin and granzyme gene expression in fresh human NK cells.
Cell Immunol
165
1995
33
44
Sheridan
W
Morstyn
G
Wolf
M
Dodds
A
Lusk
J
Maher
D
Layton
J
Green
M
Souza
L
Fox
R
Granulocyte colony-stimulating factor and neutrophil recovery after high-dose chemotherapy and autologous bone marrow transplantation.
Lancet
2
1989
891
45
Taylor
K
Jagannath
S
Spitzer
G
Spinolo
J
Tucker
S
Fogel
B
Cabanillas
F
Hagemeister
F
Souza
L
Recombinant human granulocyte colony-stimulating factor hastens granulocyte recovery after high-dose chemotherapy and autologous bone marrow transplantation in Hodgkin's disease.
J Clin Oncol
7
1989
1791
46
Nemunaitis
J
Rosenfeld
CS
Ash
R
Freedman
MH
Deeg
HJ
Appelbaum
F
Singer
JW
Flomenberg
N
Dalton
W
Elfenbein
GJ
Rifkin
R
Rubin
A
Agosti
J
Hayes
FA
Holcenberg
J
Shadduck
RK
Phase III randomized, double-blind placebo-controlled trial of rhGM-CSF following allogeneic bone marrow transplantation.
Bone Marrow Transplant
15
1995
949
47
Nemunaitis
J
Rabinowe
S
Singer
J
Bierman
P
Vose
J
Freedman
A
Onetto
N
Gillis
S
Oette
D
Gold
M
Buckner
C
Hanson
J
Ritz
J
Appelbaum
F
Armitage
J
Nadler
L
Recombinant granulocyute-macrophage colony-stimulating factor after autologous bone marrow transplantation for lymphoid cancer.
N Engl J Med
324
1991
1773
48
Tepler
I
Elias
L
Smith
JW
Hussein
M
Rosen
G
Chang
AYC
Moore
JO
Gordon
MS
Kuca
B
Beach
KJ
Loewy
JW
Garnick
MB
Kaye
JA
A randomized placebo-controlled trial of recombinant human interleukin-11 in cancer patients with severe thrombocytopenia due to chemotherapy.
Blood
87
1996
3607
49
Gordon
MS
McCaskill-Stevens
WJ
Battiato
LA
Loewy
J
Loesch
D
Breeden
E
Hoffman
R
Beach
KJ
Kuca
B
Kaye
J
Sledge
GW
A phase I trial of recombinant human interleukin 11 (neumega rhIL-11 growth factor) in women with breast cancer receiving chemotherapy.
Blood
87
1996
3615
50
Basser
RL
Rasko
JEJ
Clarke
K
Cebon
J
Green
MD
Hussein
S
Alt
C
Menchaca
D
Tomita
D
Marty
J
Fox
RM
Begley
CG
Thrombopoietic effects of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF ) in patients with advanced cancer.
Lancet
348
1996
1279
51
Basser
RL
Rasko
J
Clarke
K
Cebon
J
Green
MD
Grigg
AP
Zalcberg
J
Cohen
B
O'Byre
J
Menchaca
DM
Fox
RM
Begley
CG
Randomized, blinded, placebo-controlled phase I trial of pegylated recombinant human megakaryocyte growth and development factor with filgrastim after dose-intensive chemotherapy in patients with advanced cancer.
Blood
89
1997
3118
52
Vowels
MR
Tiedemann
K
Lam-Po-Tank R
Tucker
DP
Use of granulocyte-macrophage colony-stimulating factor in two children treated with cord blood transplantation.
Blood Cells
20
1994
249
53
Carson
WE
Ross
ME
Baiocchi
RA
Marien
MJ
Boiani
N
Grabstein
K
Caligiuri
MA
Endogenous prodution of interleukin 15 by activated human monocytes is critical for optimal production of interferon-γ by natural killer cells in vitro.
J Clin Invest
96
1995
2578
54
Doherty
TM
Seder
RA
Sher
A
Induction and regulation of IL-15 expression in murine macrophages.
J Immunol
156
1996
735
55
Nishimura
H
Hiromatsu
K
Kobayashi
N
Grabstein
KH
Paxton
R
Sugamura
K
Bluestone
JA
Yoshikai
Y
IL-15 is a novel growth factor for murine γδ T cells induced by salmonella infection.
J Immunol
156
1996
663
56
Brawerman
G
Finding the right targets.
Cell
57
1989
9
57
Brawerman
G
Determinants of messenger RNA stability.
Cell
48
1987
5
58
Shaw
G
Kamen
R
A conserved AU sequence from the 3′ untranslated region of GM-CSF mRNA mediates selective mRNA degradation.
Cell
46
1986
659
59
Caput
D
Beutler
B
Hartog
K
Thayer
R
Brown-Shirmen
S
Cerami
A
Identification of a common nucleotide sequence in the 3′-untranslated region of mRNA molecules specifying inflammatory mediators.
Proc Natl Acad Sci USA
83
1986
1670
60
Brewer
G
Ross
J
Poly (A) shortening and degradation of the 3′ A + U-rich sequences of human c-myc mRNA in a cell-free system.
Mol Cell Biol
8
1988
1697
61
Malter
J
Identification of an AUUUA-specific messenger RNA binding protein.
Science
246
1989
664
62
Brewer
G
An A+U-rich element RNA-binding factor regulates c-myc mRNA stability in vitro.
Mol Cell Biol
11
1991
2460
63
Wong
G
Witek
JS
Temple
PA
Wilkkens
KM
Leary
AC
Luxemburg
DP
Jones
SS
Brown
EL
Kay
RM
Orr
EC
Shoemaker
C
Golde
DW
Kaufman
RJ
Hewick
RM
Want
EA
Clark
SC
Human GM-CSF: Molecular cloning of the complementary DNA and purification of the natural and recombinant proteins.
Science
228
1985
810
64
Wong
G
Temple
P
Leary
A
Witek-Gianotti
J
Yang
Y
Ciarletta
A
Chung
M
Murtha
P
Kriz
R
Kaufman
R
Human CSF-1: Molecular cloning and expression of 4kb cDNA encoding the human urinary protein.
Science
235
1987
1504
65
Buzby
JS
Lee
SM
DeMaria
CT
Brewer
G
Van Winkle
P
Cairo
MS
Increased granulocyte-macrophage colony-stimulating factor mRNA instability in cord versus adult mononuclear cells is translation-dependent and associated with increased levels of A+U-rich element binding factor.
Blood
88
1996
2889
66
Jochum
C
Voth
R
Rossol
S
Buschenfelde
K
Hess
G
Will
H
Schroder
H
Steffen
R
Muller
W
Immunosuppressive function of hepatitis B antigens in vitro: Role of endoribonuclease V as one potential trans inactivator for cytokines in macrophages and human hepatoma cells.
J Virol
64
1990
1956
67
Vakalopoulou
E
Schaack
J
Shenk
T
A 32-kilodalton protein binds to AU-rich domains in the 3′ untranslated regions of rapidly degraded mRNAs.
Mol Cell Biol
11
1991
3355
68
Bohjanen
P
Petryniak
B
June
C
Thompson
C
Lindsten
T
AU RNA-binding factors differ in their binding specificities and affinities.
J Biol Chem
267
1992
6302
69
Hamilton
B
Nagy
E
Malter
J
Arrick
B
Rigby
W
Association of heterogeneous nuclear ribonucleoprotein A1 and C proteins with reiterated AUUUA sequences.
J Biol Chem
268
1993
8881
70
Levine
T
Gao
F
King
P
Andrews
L
Keene
J
Hel-N1: An autoimmune RNA-binding protein with specficity for 3′ uridylate-rich untranslated regions of growth factor mRNAs.
Mol Cell Biol
13
1993
3494
71
Nakagawa
J
Waldner
H
Meyer-Monard
S
Hofsteenge
J
Jeno
P
Moroni
C
AUH, a gene encoding and AU-specific RNA binding protein with intrinsic enoyl-CoA hydratase activity.
Proc Natl Acad Sci USA
92
1995
2051
72
Rosenberg
S
Lotze
M
Muul
L
Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer.
N Engl J Med
313
1985
1485
73
Lee
RE
Lotze
MT
Skibber
JM
Tucker
E
Bonow
RO
Ognibene
FP
Carrasquillo
JA
Shelhamer
JH
Parrillo
JE
Rosenberg
SA
Cardiorespiratory effects of immunotherapy with interleukin-2.
J Clin Oncol
7
1989
7
74
Atkins MB, Robertson M, Gordon MS, Lotze MT, Du Bois J, Ritz J, Sandler A, Edington HD, Sherman ML: Phase I evaluation of intravenous recombinant human interleukin-12 (RHIL-12) in patients with advanced malignancies. Proc Am Soc Clin Oncol 15:270, 1996 (abstr)
75
Ross
ME
Caligiuri
MA
Cytokine-induced apoptosis of human natural killer (NK) cells identifies a novel mechanism to regulate the innate immune response.
Blood
89
1997
910
Sign in via your Institution