Interleukin-10 (IL-10) selectively inhibited lipopolysaccharide (LPS)-induced chemoattractant cytokine gene expression: levels of IP-10 mRNA were markedly suppressed in IL-10–treated mouse peritoneal macrophages, whereas the expression of the RANTES mRNA was only modestly reduced. IL-10 inhibited IP-10 mRNA accumulation by reducing IP-10 gene transcription as demonstrated by nuclear run-on analysis. Interestingly, the ability of IL-10 to inhibit expression of IP-10 was dependent on the inducing stimulus; IL-10 did not suppress interferon γ (IFNγ)- or IFNβ-stimulated IP-10 transcription or mRNA accumulation. These results suggested that IL-10 might act indirectly to suppress IP-10 expression by inhibiting LPS-induced class I IFN production. This hypothesis was supported by the following observations. First, LPS-induced IP-10 mRNA expression was blocked in cells cotreated with cycloheximide. Second, IL-10 inhibited the production of IFN/β-mediated antiviral activity. Finally, the IL-10–mediated suppression of LPS-stimulated IP-10 production could be rescued by cotreatment with IFNβ.

MONONUCLEAR phagocytes play an important role in the orchestration of acute and chronic inflammation.1-3 The behavioral potential of macrophages during an inflammatory process is determined, in part, by multiple signals encountered in the tissue microenvironment. These may include bacterial cell wall products such as lipopolysaccharide (LPS) and secreted cytokines from antigen-stimulated T cells such as interferon γ (IFNγ). Because the inflammatory functions of macrophages are potentially damaging to normal tissue, precise control of the response is necessary and is frequently provided by anti-inflammatory cytokines such as interleukin-10 (IL-10).

IL-10 has been shown to have profound effects on monocytes and macrophages, causing inhibition of a number of LPS-induced proinflammatory cytokines such as IL-1α, IL-6, IL-8, and tumor necrosis factor α (TNFα).4-8 The molecular mechanisms through which this inhibitory function is achieved have been the subject of multiple studies, the results of which are diverse.9-18 Thus, IL-10 has been reported to inhibit stimulus-induced monocyte/macrophage gene expression by blocking transcription, by altering the stability of mRNAs, and by reducing the translation of mRNAs.

This mechanistic heterogeneity could reflect differences in the signaling events induced by the stimuli that promote their expression. To determine if differential signaling from distinct inducing stimuli might account for differential sensitivity to IL-10, we have chosen to evaluate the mechanisms involved in IL-10–mediated control of the gene encoding the chemoattractant cytokine (chemokine) IP-10. The IP-10 gene is an appealing model for this purpose, because the mechanisms involved in controlling induced expression are well characterized.19-23 More importantly, the gene can be induced in macrophages in response to multiple stimuli, including virus, poly IC, LPS, IFNγ, and IFNα/β.19 22-26 Our findings demonstrate that IL-10 suppresses IP-10 expression in a stimulus-dependent fashion: response to LPS is sensitive, whereas that to IFNγ or IFNβ is unaffected by IL-10. The suppression of IP-10 appears to be indirect and depends on the immediate inhibition of LPS-induced type I IFN.

Reagents.

Brewer’s Thioglycollate broth (TG) was purchased from Difco Laboratories (Detroit, MI). LPS prepared from the Escherichia coliserotype 0111:B4 was purchased from Sigma Chemical Co (St Louis, MO). Recombinant IFNγ, RPMI 1640, antibiotics, and glutamine were purchased from GIBCO BRL Life Technologies (Gaithersburg, MD). IFNβ and IL-10 were obtained from Genzyme Inc (Cambridge, MA). Fetal bovine serum (FBS) was obtained from Hyclone (Logan, UT) and was heat-inactivated before use. All cell culture reagents were specified to be endotoxin free. Cesium chloride, guanidine thiocyanate, agarose, sodium dodecyl sulfate (SDS), Tris, proteinase K, RNase-free DNase, and random priming kits were purchased from Boehringer Mannheim Corp (Indianapolis, IN). Formamide was obtained from US Biochemical (Cleveland, OH). Dextran sulfate was obtained from Pharmacia (Uppsala, Sweden). Nylon transfer membrane was purchased from Micron Separations Inc (Westborough, MA). Dupont NEN Research Products (Boston, MA) was the source of α[32P]-dCTP and α[32P]-UTP.

Mice.

Specific pathogen-free, inbred C57BL/6 mice, 9 to 12 weeks of age, were purchased from Jackson Laboratories (Bar Harbor, ME) and housed in microisolator cages with autoclaved food and bedding to minimize exposure to viral and microbial pathogens and to ensure that the degree of spontaneous activation of tissue macrophages would be minimal.27 

Cell culture.

TG-elicited macrophages were obtained as reported previously.28 29 Peritoneal lavage was performed using 10 mL of cold Hanks’ balanced salt solution (HBSS) containing 10 U/mL heparin. Macrophages were plated in 100- or 150-mm tissue culture dishes, incubated for 2 hours at 37°C in an atmosphere of 5% CO2, and then washed three times with HBSS to remove nonadherent cells. The macrophages were cultured overnight in RPMI 1640 at 37°C in 5% CO2 and then cultured in the presence or absence of stimuli for the indicated times.

Preparation of plasmid DNA.

The plasmids encoding genes IP-10, RANTES, α-tubulin, and GAPDH were as described previously.30,31 Methods for plasmid preparation were as described by Sambrook et al.32 

Preparation of RNA and Northern hybridization analysis.

Total cellular RNA was prepared by the guanidine thiocyanate-cesium chloride method.33 Equal amounts of RNA (10 μg) were used in each lane of the gel. The RNA was denatured, separated by electrophoresis in a 1% agarose-formaldehyde gel, and transferred to a nylon membrane as previously described.28 29 The blots were prehybridized for 12 to 24 hours at 42°C in 50% formamide, 1% SDS, 5× SSC, 1× Denhardt’s (0.02% Ficoll, 0.02% bovine serum albumin [BSA], and 0.02% polyvinylpyrrolidone), 0.25 mg/mL denatured herring testis DNA, and 50 mmol/L sodium phosphate buffer, pH 6.5. Hybridization was performed at 42°C for 18 hours with 1 × 107 cpm of denatured plasmid DNA containing appropriate specific cDNA inserts. The filters were rinsed with a solution of 0.1% SDS-0.2× SSC, washed at 42°C for 1 hour, and washed at 65°C for 15 minutes. The blots were dried and exposed by using XAR-5 x-ray film (Eastmann Kodak Co, Rochester, NY) with Dupont (Wilmington, DE) Cronex Lightening Plus intensifying screens at −70°C. Blots were quantified by phosphoresence analysis using an instrument from Molecular Dynamics (Sunnyvale, CA).

Nuclear transcription assay.

Cultures of 5 × 107 macrophages were treated as indicated in the text and nuclei isolated as described previously.24,34 Transcription initiated in intact cells was allowed to complete in the presence of α-[32P]-UTP and the RNA was isolated and hybridized to slot-blotted plasmids containing specific cDNA (7 μg DNA/slot) essentially as described elsewhere.24 34 Blots were hybridized for 72 hours and exposed to x-ray film for 2 to 4 days. The α-tubulin gene was used as an internal standard. The expression of specific transcripts was quantified by phosphorimage analysis. Numerical values for specific transcripts were normalized to α-tubulin transcript level in the same sample. This ratio in untreated samples was arbitrarily set to unity. Experimental values are presented as fold induction relative to untreated samples.

IFNα/β-mediated antiviral activity.

Macrophages were stimulated in complete medium for 4 hours with LPS in the presence or absence of IL-10. Supernatant medium was collected, centrifuged to pellet residual cells and cell debris, and used to measure antiviral activity as described previously.35 Mouse embryo fibroblasts (MEF) were plated at a density of 5 × 104 per well in 24-well plates. Cultures were treated with medium alone or medium containing various dilutions of macrophage culture supernatants or known quantities of IFNβ. After 24 hours, MEF cultures were washed with fresh medium, infected with endomyocarditis (EMC) virus (multiplicity of infection [MOI] = 2; gift of Dr Robert Silverman, Cleveland Clinic Foundation, Cleveland, OH) and incubation continued for an additional 24 hours. The cell controls included MEF cells cultured with supernatants alone or with EMC infection. Cell viability was measured by staining wells with neutral red in PBS and elution in 50% ethanol in 0.1 mol/L NaH2PO4, and the absorbance at 540 nm was determined. The results are presented as the percentage of protection calculated as follows: [A540(sample) − A540(virus control)]/[A540(cell control) − A540(virus control)] × 100.

IL-10 selectively inhibits expression of LPS-induced chemokine mRNAs.

Previous work from multiple laboratories has demonstrated that IL-10 is a potent suppressor of induced gene expression in mononuclear phagocytes.4-8 The mechanisms through which such inhibitory effect is achieved are diverse.9-18 To distinguish the importance of stimulus specificity in determining the mechanisms involved in IL-10–mediated suppression, we have examined the effects of IL-10 on the chemokine gene IP-10, the control of which has been extensively studied.20-23 Peritoneal macrophages were treated with LPS for 2 hours in the presence or absence of IL-10, and expression of mRNAs encoding IP-10 or RANTES was analyzed by Northern hybridization. In agreement with previous results, LPS treatment stimulated strong expression of IP-10 mRNA, which was markedly reduced (>80% as quantified by phosphorimage analysis) in macrophages treated simultaneously with IL-10 (Fig 1). This effect was gene selective, because levels of RANTES mRNA were only modestly reduced (<25%).

Fig. 1.

IL-10 selectively suppresses LPS-induced chemokine gene expression. TG-elicited macrophages (1 × 107) were treated with LPS (10 ng/mL) in the absence or presence of IL-10 (25 ng/mL) for 2 hours. Total RNA was prepared and the levels of IP-10, RANTES, and GAPDH mRNA were analyzed using northern hybridization. Similar results were obtained in three separate experiments.

Fig. 1.

IL-10 selectively suppresses LPS-induced chemokine gene expression. TG-elicited macrophages (1 × 107) were treated with LPS (10 ng/mL) in the absence or presence of IL-10 (25 ng/mL) for 2 hours. Total RNA was prepared and the levels of IP-10, RANTES, and GAPDH mRNA were analyzed using northern hybridization. Similar results were obtained in three separate experiments.

Close modal

The time dependence for IL-10–mediated suppression of LPS-induced IP-10 mRNA expression was determined in macrophages treated for various times (Fig 2). LPS-stimulated expression of IP-10 mRNA was transient: the peak levels were obtained within 4 hours, declined markedly by 8 hours, and returned to nearly baseline after 16 hours of stimulation. IL-10 suppressed the production of IP-10 mRNA, although the inhibition was not evident until 2 hours of IL-10 exposure.

Fig. 2.

Time dependence of IL-10–mediated suppression of LPS-induced IP-10 mRNA expression. (A) TG-elicited macrophages were treated with LPS (10 ng/mL) in the absence or presence of IL-10 (25 ng/mL) for the indicated times. Total RNA was prepared and levels of IP-10 and GAPDH mRNA were analyzed by northern hybridization. (B) The levels of specific mRNA on the blot shown in (A) were quantified by phosphorimage analysis. Levels of IP-10 mRNA in each sample are normalized for the levels of GAPDH mRNA. Similar results were obtained in two separate experiments.

Fig. 2.

Time dependence of IL-10–mediated suppression of LPS-induced IP-10 mRNA expression. (A) TG-elicited macrophages were treated with LPS (10 ng/mL) in the absence or presence of IL-10 (25 ng/mL) for the indicated times. Total RNA was prepared and levels of IP-10 and GAPDH mRNA were analyzed by northern hybridization. (B) The levels of specific mRNA on the blot shown in (A) were quantified by phosphorimage analysis. Levels of IP-10 mRNA in each sample are normalized for the levels of GAPDH mRNA. Similar results were obtained in two separate experiments.

Close modal
Mechanisms of IL-10–mediated suppression of IP-10 mRNA expression.

LPS-induced IP-10 mRNA expression is mediated by increases in gene transcription.20-22 To determine if the suppression of LPS-induced IP-10 mRNA by IL-10 involved inhibition of transcription, nuclear run-on experiments were performed. Cultures of peritoneal macrophages were treated with LPS for various times in the absence or presence of IL-10, the nuclei were harvested, and the RNA transcripts initiated in vivo were allowed to elongate in vitro in the presence of32P-UTP. The radiolabeled RNA products were hybridized to slot-blotted cDNAs encoding IP-10 and α-tubulin and the resulting blots were quantified by phosphorimage analysis (Fig 3). The transcriptional activity of the IP-10 gene was elevated within 2 hours and appeared to be optimally active after stimulation with LPS for 4 hours. This LPS-dependent stimulation of transcription of the IP-10 gene was almost fully suppressed in cells that had been cotreated with IL-10.

Fig. 3.

IP-10 mRNA expression is controlled at the level of transcription. (A) TG-elicited macrophages were treated with LPS (10 ng/mL) for various times in the absence or presence of IL-10 (25 ng/mL) before isolation of nuclei and analysis of transcription by nuclear run-on. Radiolabeled nuclear RNA was hybridized with nylon membranes containing equivalent amounts of denatured plasmid DNA encoding IP-10 and tubulin (top panel). (B) Slot blots shown in the top panel were quantified by phosphorimage analysis and levels of IP-10 transcript normalized for levels of tubulin. Similar results were obtained in three separate experiments.

Fig. 3.

IP-10 mRNA expression is controlled at the level of transcription. (A) TG-elicited macrophages were treated with LPS (10 ng/mL) for various times in the absence or presence of IL-10 (25 ng/mL) before isolation of nuclei and analysis of transcription by nuclear run-on. Radiolabeled nuclear RNA was hybridized with nylon membranes containing equivalent amounts of denatured plasmid DNA encoding IP-10 and tubulin (top panel). (B) Slot blots shown in the top panel were quantified by phosphorimage analysis and levels of IP-10 transcript normalized for levels of tubulin. Similar results were obtained in three separate experiments.

Close modal
IL-10–mediated suppression of IP-10 mRNA is stimulus-dependent.

IFNγ and IFNβ are also important stimuli of IP-10 gene expression.22,24 36 To determine whether the effect of IL-10 on IP-10 expression varies with the inducing stimulus, peritoneal macrophages were incubated with IFNγ or IFNβ in the absence or presence of IL-10 for various times and IP-10 mRNA levels were measured by Northern hybridization and phosphorimage analysis (Fig 4). IFNγ-stimulated IP-10 mRNA expression reached maximum levels within 2 hours, whereas IFNβ-induced IP-10 mRNA levels were maximal at 4 hours. Neither IFNγ nor IFNβ stimulated the expression of the RANTES gene (data not shown). In contrast to the marked suppression of LPS-induced IP-10 levels seen in Figs 1 and 2, IL-10 had no significant effect on IP-10 mRNA levels stimulated by either IFNγ or IFNβ. This difference was confirmed at the level of transcription; IP-10 gene transcription induced by either IFNβ or IFNγ was insensitive to IL-10 treatment, whereas transcription in response to LPS was selectively inhibited (Fig 5).

Fig. 4.

The suppressive action of IL-10 is stimulus-dependent. TG-elicited macrophages were treated with IFNγ (50 U/mL; A) or IFNβ (500 U/mL; B) in the absence or presence of IL-10 (25 ng/mL) for the indicated times. Total RNA was prepared and IP-10 and GAPDH mRNA were analyzed using northern hybridization. Levels of expression in each lane were quantified using phosphorimage analysis. The data presented for IP-10 mRNA are normalized to the level of GAPDH in each sample. Similar results were obtained in two separate experiments.

Fig. 4.

The suppressive action of IL-10 is stimulus-dependent. TG-elicited macrophages were treated with IFNγ (50 U/mL; A) or IFNβ (500 U/mL; B) in the absence or presence of IL-10 (25 ng/mL) for the indicated times. Total RNA was prepared and IP-10 and GAPDH mRNA were analyzed using northern hybridization. Levels of expression in each lane were quantified using phosphorimage analysis. The data presented for IP-10 mRNA are normalized to the level of GAPDH in each sample. Similar results were obtained in two separate experiments.

Close modal
Fig. 5.

IL-10 selectively inhibits transcription of the IP-10 gene. TG-elicted macrophages (1 × 107 cells/sample) were stimulated with LPS (10 ng/mL) or IFNβ (500 U/mL) in the presence or absence of IL-10 (25 ng/mL) for 2 hours. In a separate experiment, macrophages were treated with LPS or IFNγ plus or minus IL-10. Nuclei were prepared and used in nuclear run-on assay as described in the legend to Fig 3. Similar results were obtained in two separate experiments.

Fig. 5.

IL-10 selectively inhibits transcription of the IP-10 gene. TG-elicted macrophages (1 × 107 cells/sample) were stimulated with LPS (10 ng/mL) or IFNβ (500 U/mL) in the presence or absence of IL-10 (25 ng/mL) for 2 hours. In a separate experiment, macrophages were treated with LPS or IFNγ plus or minus IL-10. Nuclei were prepared and used in nuclear run-on assay as described in the legend to Fig 3. Similar results were obtained in two separate experiments.

Close modal
The inhibition of LPS-induced IP-10 expression by IL-10 is mediated by the suppression of LPS-induced IFNβ production.

The finding that IP-10 expression induced by IFNγ or IFNβ was insensitive to inhibition by IL-10 suggested the possibility that the effects of IL-10 on LPS-induced IP-10 expression might involve an indirect mechanism. In a previous report we observed that the ability of LPS to stimulate transcription from the promoter of the IP-10 gene was blocked by including antibody against type I IFNs in the culture medium.21 Thus, IFNβ, induced in response to LPS, might be the direct mediator of IP-10 expression and the immediate target of IL-10 suppression; IL-10 could inhibit IFNβ expression and indirectly the LPS-induced expression of IP-10. To test this possibility, we have performed three distinct experiments. First, we have determined if IP-10 expression in response to LPS depends on intermediate protein synthesis. Macrophages were treated with LPS in the presence or absence of cycloheximide for 4 hours and the total RNA was then analyzed for IP-10 mRNA content. Consistent with our hypothesis and previous results, LPS-induced IP-10 expression was blocked in cells treated with cycloheximide (Fig 6).

Fig. 6.

LPS-induced IP-10 mRNA expression is dependent on protein synthesis. TG-elicited macrophages (1 × 107 cells/sample) were treated with LPS (10 ng/mL) in the absence or presence of cycloheximide (1 μg/mL) for 4 hours before measurement of IP-10 and GAPDH mRNA levels by northern hybridization and phosphorimage analysis as described in the legend to Fig 2. Similar results were obtained in two separate experiments.

Fig. 6.

LPS-induced IP-10 mRNA expression is dependent on protein synthesis. TG-elicited macrophages (1 × 107 cells/sample) were treated with LPS (10 ng/mL) in the absence or presence of cycloheximide (1 μg/mL) for 4 hours before measurement of IP-10 and GAPDH mRNA levels by northern hybridization and phosphorimage analysis as described in the legend to Fig 2. Similar results were obtained in two separate experiments.

Close modal

In the second experiment we determined if LPS-induced IFNβ is sensitive to suppression by IL-10. Macrophages were treated with LPS in the presence or absence of IL-10 for 4 hours. Culture supernatants were harvested and the IFNβ content was measured on the basis of IFNβ-dependent antiviral activity (Table1). LPS stimulated significant antiviral activity in two separate experiments, and this was suppressed by 75% when IL-10 was included in the culture medium. The antiviral activity was identifiable as IFNβ on the basis of complete neutralization with antibody specific for type I mouse IFNs (data not shown).

Table 1.

IL-10 Inhibits LPS-Induced Production of Type I IFN

Treatment Protection (%) Inhibition (%)
Experiment no. 1  
 None  0  NA  
 LPS  51.3  
 LPS + IL-10  12.8  75  
Experiment no. 2 
 None  0  NA  
 LPS  20.1  
 LPS + IL-10  5.8  71 
Treatment Protection (%) Inhibition (%)
Experiment no. 1  
 None  0  NA  
 LPS  51.3  
 LPS + IL-10  12.8  75  
Experiment no. 2 
 None  0  NA  
 LPS  20.1  
 LPS + IL-10  5.8  71 

TG-elicited peritoneal macrophages (1 × 107) were stimulated with LPS (10 ng/mL) without or with IL-10 (25 ng/mL) for 4 hours. Supernatants were harvested and used to pretreat cultures of MEF cells for 24 hours before infection with EMC virus at an MOI of 2. Control cultures were treated with supernatant but not infected with virus. After 24 hours, MEF cell viability was assayed. The percentage of protection was calculated as described in Materials and Methods. Antiviral activity was completely inhibited by anti-IFNα/β antibody (data not shown).

Abbreviation: NA, not applicable.

The final experiment was to determine if IL-10–mediated suppression of LPS-induced IP-10 could be rescued by inclusion of IFNβ in the culture medium. Macrophages were stimulated with LPS or LPS and IFNβ in the absence or presence of IL-10 for 4 hours before determination of IP-10 mRNA levels by Northern hybridization. As expected, LPS stimulated strong expression of IP-10, which was suppressed in this experiment by approximately 50% in the presence of IL-10 (Fig 7). Inclusion of IFNβ in the medium did not increase the levels of IP-10 mRNA but did protect against IL-10–mediated suppression.

Fig. 7.

IFNβ rescues IL-10–suppressed IP-10 mRNA expression. TG-elicited macrophages (1 × 107 cells/sample) were treated with LPS (10 ng/mL), IFNβ (500 U/mL), or IL-10 (25 ng/mL) as indicated for 4 hours before measurement of IP-10 and GAPDH mRNA levels using northern hybridization and phosphorimage analysis as described in the legend to Fig 2. Similar results were obtained in two separate experiments.

Fig. 7.

IFNβ rescues IL-10–suppressed IP-10 mRNA expression. TG-elicited macrophages (1 × 107 cells/sample) were treated with LPS (10 ng/mL), IFNβ (500 U/mL), or IL-10 (25 ng/mL) as indicated for 4 hours before measurement of IP-10 and GAPDH mRNA levels using northern hybridization and phosphorimage analysis as described in the legend to Fig 2. Similar results were obtained in two separate experiments.

Close modal

Multiple studies have demonstrated that IL-10 suppresses cytokine production in various cell types, although the mechanisms involved appear to be diverse. Inhibition may result from decreased gene transcription, reduction in specific mRNA stability, and/or reduced mRNA translation.7,9,10,12,14-16,18,37 Which of these different mechanisms participate is likely to depend on the signaling pathway(s) that controls expression. To evaluate this problem we elected to examine mechanisms involved in IL-10–mediated suppression of a chemokine gene (IP-10) known to be induced in response to several stimuli that use distinct signaling pathways.23-25 The results demonstrate that IL-10 inhibits IP-10 gene transcription through a mechanism that depends on the stimulus. The stimulus selectivity is a result of two related events: (1) LPS-induced IP-10 expression depends on the intermediate induction of class I IFNs and (2) LPS-induced expression of class I IFNs is suppressed by IL-10. These conclusions are supported by the following experimental observations. (1) IL-10 selectively reduces LPS-induced but not IFNγ- or IFNβ-induced IP-10 mRNA levels. (2) Nuclear run-on analysis indicates that LPS-stimulated IP-10 transcription is selectively suppresssed by IL-10. (3) LPS-induced IP-10 expression is blocked by inhibitors of protein synthesis. (4) Antibodies to class I IFNs can block LPS-stimulated transcription from the IP-10 promoter.21 (5) Production of LPS-induced, IFNβ-mediated antiviral activity is suppressed by IL-10. (6) IFNβ treatment rescues IL-10–mediated suppression of LPS-induced IP-10 mRNA.

LPS is known to be a potent inducer of IL-10 in macrophages.38,39 IL-10 produced under such conditions may act in both autocrine and paracrine fashion to provide negative feedback regulation of the LPS-induced inflammatory response. Including antibody to IL-10 in cultures of LPS-treated macrophages enhances some LPS-induced gene expression and, indeed, the autocrine/paracrine action of IL-10 appears to be a component of LPS-induced tolerance to second endotoxin exposure.40 41 

Several reports have shown that gene transcription can be a mechanistic target for the suppressive function of IL-10, and our results demonstrating that IL-10 suppresses LPS-stimulated transcription of the IP-10 gene provide further confirmation.10,14 On the basis of electrophoretic mobility shift assays, Wang et al42showed that IL-10 could inhibit the LPS-induced nuclear localization of NFκB in human monocytes and that NFκB is likely to be an important transcription factor controlling the expression of the IP-10 gene.21,26,43-45 The marked sensitivity to IL-10 of LPS-induced but not IFN-induced IP-10 expression suggests the possibility that IL-10 might differentially target the intracellular signalling pathways used by these agents: IP-10 induced by LPS might be more dependent on activation of NFκB than IP-10 induced by IFN and could, therefore, exhibit greater sensitivity to the action of IL-10. Our own studies indicate that IL-10 does not block the activation of NFκB in macrophages stimulated with LPS and thus does not support NFκB as a target of IL-10 (data not shown). Rather, the data indicate that the action of IL-10 on LPS-induced IP-10 is indirect. In support of this, the induction of IP-10 by LPS is apparently indirect; inhibition of protein synthesis blocked the expression of IP-10 when LPS was the stimulus. Furthermore, in a previous report, we observed that neutralizing antibody against IFNα/β could block the ability of LPS to stimulate reporter gene expression driven by the IP-10 promoter.21 LPS is well known to induce class I IFNs,46,47 and our findings now show that LPS-induced class I IFN production is strongly suppressed by IL-10. Finally, inclusion of IFNβ in the culture medium rescues IL-10–suppressed IP-10 expression. Thus, IL-10 acts to suppress the intermediate expression of IFNβ, the result of which is indirect inhibition of IP-10 gene transcription.24 

Many studies of IL-10–mediated suppression of gene expression have implicated accelerated mRNA decay as an important contributing mechanism. mRNAs that exhibit shortened half lives in IL-10–treated cells include TNFα, IL-1α, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-10, IL-6, and the chemokine genes macrophage inflammatory protein-1α (MIP-1α), MIP-1β, and IL-8.9,11,14,15,18 In previous work, we have also observed significant effects of IL-10 on mRNA stability.48 The intracellular pathway(s) through which IL-10 enhances mRNA degradation is currently unknown. AU-rich sequence elements (AREs) in the 3′-untranslated regions (3′UTRs) of short-lived mRNAs have been shown to confer accelerated decay upon stable mRNAs,49-52 and it is noteworthy that mice bearing a mutant TNFα allele, in which the ARE in the 3′-UTR has been altered, have lost regulation of TNFα expression in vivo in response to IL-10, suggesting that this motif may be involved.53 ARE sequences are present in the 3′UTRs of a variety of cytokine mRNAs that are sensitive to suppression by IL-10 (ie, GM-CSF, TNFα, IL-1α, IL-1β, MIP-1α, and KC) but are absent in RANTES mRNA that is insenstive to suppression by IL-1054-61 (see Fig 1). The IFNβ mRNA sequence contains an ARE motif that has been reported to be important in regulating levels of mRNA under various circumstances.62-64 Thus, it is possible that the IL-10–mediated suppression of LPS-induced IP-10 gene transcription depends on destabilization of IFNβ mRNA through a mechanism that may involve the ARE motif.

The IL-10–induced signaling pathways that mediate its inhibitory effects remain incompletely defined. The IL-10 receptor is most closely related to the receptors for class I and class II IFNs. Consistent with this relationship, IL-10 treatment of T cells and monocytes stimulates tyrosine phosphorylation of the Janus family protein kinases, TYK2 and Jak1, and the phosphorylation and nuclear translocation of STAT1α and STAT3.65-67 A recent report indicates that STAT3 docking sites on the intracellular domain of the IL-10 receptor are required for IL-10–mediated suppression, although activation of STAT3 itself may not be necessary.68 In addition, IL-10 has been reported to suppress LPS-induced protein tyrosine phosphorylation.69 However, the specific roles for these various intracellular signaling events will require additional analysis.

Supported by US Public Health Services Grants No. CA39621 and CA62220.

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

1
Adams
DO
Hamilton
TA
The cell biology of macrophage activation [review].
Annu Rev Immunol
2
1984
283
2
Adams
DO
Hamilton
TA
Macrophages as destructive cells in host defense
Inflammation: Basic Principles and Clinical Correlates.
Gallin
JI
Goldstein
IM
Snyderman
R
1992
637
Raven
New York, NY
3
Nathan
CF
Cohn
ZA
Cellular components of inflammation: Monocytes and macrophages
Textbook of Rheumatology.
Kelly
W
Harris
E
Ruddy
S
Hedge
R
1995
144
Saunders
Philadelphia, PA
4
Bogdan
C
Vodovotz
Y
Nathan
C
Macrophage deactivation by interleukin 10.
J Exp Med
174
1991
1549
5
Moore
KW
O’Garra
A
De Waal Malefyt
R
Vieira
P
Mosmann
TR
interleukin-10.
Annu Rev Immunol
11
1993
165
6
Ho
AS
Moore
KW
Interleukin-10 and its receptor.
Ther Immunol
1
1994
173
7
Fiorentino
DF
Zlotnik
A
Mosmann
TR
Howard
M
O’Garra
A
IL-10 inhibits cytokine production by activated macrophages.
J Immunol
11
1991
3815
8
Ohmori
Y
Hamilton
TA
Regulation of macrophage gene expression by T cell derived lymphokines.
Pharmacol Ther
63
1994
235
9
Bogdan
C
Paik
J
Vodovotz
Y
Nathan
C
Contrasting mechanisms for suppression of macrophage cytokine release by transforming growth factor-β and interleukin-10.
J Biol Chem
267
1992
23301
10
Wang
P
Wu
P
Siegel
MI
Egan
RW
Billah
MM
IL-10 inhibits transcription of cytokine genes in human peripheral blood mononuclear cells.
J Immunol
153
1994
811
11
Kasama
T
Strieter
RM
Lukacs
NW
Burdick
MD
Kunkel
SL
Regulation of neutrophil-derived chemokine expression by IL-10.
J Immunol
152
1994
3559
12
Shrikant
P
Weber
E
Jilling
T
Benveniste
EN
Intercellular adhesion molecule-1 gene expression by glial cells. Differential mechanisms of inhibition by IL-10 and IL-6.
J Immunol
155
1995
1489
13
Seitz
M
Loetscher
P
Dewald
B
Towbin
H
Gallati
H
Baggiolini
M
Interleukin-10 differentially regulates cytokine inhibitor and chemokine release from blood mononuclear cells and fibroblasts.
Eur J Immunol
25
1995
1129
14
Berkman
N
John
M
Roesems
G
Jose
PJ
Barnes
PJ
Chung
KF
Inhibition of macrophage inflammatory protein-1α expression by IL-10. Differential sensitivities in human blood monocytes and alveolar macrophages.
J Immunol
155
1995
4412
15
Takeshita
S
Gage
JR
Kishimoto
T
Vredevoe
DL
Martı́nez-Maza
O
Differential regulation of IL-6 gene transcription and expression by IL-4 and IL-10 in human monocytic cell lines.
J Immunol
156
1996
2591
16
Song
S
Ling-Hu
H
Roebuck
KA
Rabbi
MF
Donnelly
RP
Finnegan
A
Interleukin-10 inhibits interferon-γ–induced intercellular adhesion molecule-1 gene transcription in human monocytes.
Blood
89
1997
4461
17
Wang
P
Wu
P
Anthes
JC
Siegel
MI
Egan
RW
Billah
MM
Interleukin-10 inhibits interleukin-8 production in human neutrophils.
Blood
83
1994
2678
18
Brown
CY
Lagnado
CA
Vadas
MA
Goodall
GJ
Differential regulation of the stability of cytokine mRNAs in lipopolysaccharide-activated blood monocytes in response to interleukin-10.
J Biol Chem
271
1996
20108
19
Vanguri
P
Farber
JM
IFN and virus-inducible expression of an immediate early gene, crg-2/IP-10, and a delayed gene, I-Aa, in astrocytes and microglia.
J Immunol
152
1994
1411
20
Shin
HS
Drysdale
B-E
Shin
ML
Noble
PW
Fisher
SN
Paznekas
WA
Definition of a lipopolysaccharide-responsive element in the 5′-flanking regions of MuRantes and crg-2.
Mol Cell Biol
14
1994
2914
21
Ohmori
Y
Hamilton
TA
Cooperative interaction between interferon (IFN) stimulus response element and kB sequence motifs controls IFNγ- and lipopolysaccharide-stimulated transcription from the murine IP-10 promoter.
J Biol Chem
268
1993
6677
22
Luster
AD
Unkeless
JC
Ravetch
JB
Gamma interferon transcriptionally regulates an early response gene containing homology to platelet proteins.
Nature
315
1985
672
23
Nazar
ASMI
Cheng
GH
Shin
HS
Brothers
PN
Dhib-Jalbut
S
Shin
ML
Vanguri
P
Induction of IP-10 chemokine promoter by measles virus: Comparison with interferon-gamma shows the use of the same response element but with differential DNA-protein binding profiles.
J Neuroimmunol
77
1997
116
24
Hamilton
TA
Bredon
N
Ohmori
Y
Tannenbaum
CS
IFN-gamma and IFN-beta independently stimulate the expression of lipopolysaccharide-inducible genes in murine peritoneal macrophages.
J Immunol
142
1989
2325
25
Ohmori
Y
Hamilton
TA
A macrophage LPS-inducible early gene encodes the murine homologue of IP-10.
Biochem Biophys Res Commun
3
1990
1261
26
Wu
C
Ohmori
Y
Bandyopadhyay
S
Sen
G
Hamilton
T
Interferon-stimulated response element and NFκB sites cooperate to regulate double-stranded RNA-induced transcription of the IP-10 gene.
J Interferon Res
6
1994
357
27
Meltzer
MS
Tumoricidal response in vitro of peritoneal macrophages from conventionally housed and germ-free nude mice.
Cell Immunol
22
1976
176
28
Introna
M
Hamilton
TA
Kaufman
RE
Adams
DO
Bast
RC
Jr
Treatment of murine peritoneal macrophages with bacterial lipopolysaccharide alters expression of c-fos and c-myc oncogenes.
J Immunol
8
1986
2711
29
Tannenbaum
CS
Koerner
TJ
Jansen
MM
Hamilton
TA
Characterization of lipopolysaccharide-induced macrophage gene expression.
J Immunol
140
1988
3640
30
Ohmori
Y
Hamilton
TA
IFNγ selectively inhibits lipopolysaccharide-inducible JE/monocyte chemoattractant protein-1 and KC/gro/melanoma growth-stimulating activity gene expression in mouse peritoneal macrophages.
J Immunol
5
1994
2204
31
Tannenbaum
CS
Wicker
N
Armstrong
D
Tubbs
R
Finke
J
Bukowski
RM
Hamilton
TA
Cytokine and chemokine expression in tumors of mice receiving systemic therapy with IL-12.
J Immunol
156
1996
693
32
Sambrook
J
Fritsch
EF
Maniatis
T
Molecular Cloning: A Laboratory Manual.
1989
Cold Spring Harbor Laboratory
Cold Spring Harbor, NY
33
Chirgwin
JM
Pryzbyla
RJ
MacDonald
RJ
Rutter
WJ
Isolation of biologically active RNA from sources enriched in ribonuclease.
Biochemistry
18
1979
5295
34
Koerner
TJ
Hamilton
TA
Introna
M
Tannenbaum
CS
Bast
RC
Jr
Adams
DO
The early competence genes JE and KC are differentially regulated in murine peritoneal macrophages in response to lipopolysaccharide.
Biochem Biophys Res Commun
3
1987
969
35
Johnston
MD
Finter
NB
Young
PA
Dye uptake method for assay of interferon activity.
Methods Enzymol
78
1981
394
(part A)
36
Vanguri
P
Farber
JM
Identification of crg-2, an interferon-inducible mRNA predicted to encode a murine monokine.
J Biol Chem
265
1990
15049
37
Kasama
T
Strieter
RM
Standiford
TJ
Burdick
MD
Kunkel
SL
Expression and regulation of human neutrophil-derived macrophage inflammatory protein 1α.
J Exp Med
178
1993
63
38
Barsig
J
Kusters
S
Vogt
K
Volk
HD
Tiegs
G
Wendel
A
Lipopolysaccharide-induced interleukin-10 in mice: Role of endogenous tumor necrosis factor-alpha.
Eur J Immunol
25
1995
2888
39
Salkowski
CA
Detore
GR
Vogel
SN
Lipopolysaccharide and monophosphoryl lipid A differentially regulate interleukin-12, gamma interferon, and interleukin-10 mRNA production in murine macrophages.
Infect Immun
65
1997
3239
40
Randow
F
Syrbe
U
Meisel
C
Krausch
D
Zuckermann
H
Platzer
C
Volk
HD
Mechanism of endotoxin desensitization: Involvement of interleukin 10 and transforming growth factor beta.
J Exp Med
181
1995
1887
41
Shnyra
A
Brewington
R
Alipio
A
Amura
C
Morrison
DC
Reprogramming of lipopolysaccharide-primed macrophages is controlled by a counterbalanced production of IL-10 and IL-12.
J Immunol
160
1998
3729
42
Wang
P
Wu
P
Siegel
MI
Egan
RW
Billah
MM
Interleukin (IL)-10 inhibits nuclear factor κ B (NFκB) activation in human monocytes. IL-10 and IL-4 suppress cytokine synthesis by different mechanisms.
J Biol Chem
270
1995
9558
43
Ohmori
Y
Tebo
J
Nedospasov
S
Hamilton
TA
κB binding activity in a murine macrophage-like cell line. Sequence-specific differences in κB binding and transcriptional activation functions.
J Biol Chem
269
1994
17684
44
Tebo
JM
Chaoqun
W
Ohmori
Y
Hamilton
TA
Murine inhibitory protein-κBα negatively regulates κB-dependent transcription in lipopolysaccharide-stimulated RAW264.7 macrophages.
J Immunol
10
1994
4713
45
Ohmori
Y
Hamilton
TA
The interferon stimulated response element and a κB site mediate synergistic induction of murine IP-10 gene transcription by IFNγ and TNFα.
J Immunol
154
1995
5235
46
Fujihara
M
Ito
N
Pace
JL
Watanabe
Y
Russell
SW
Suzuki
T
Role of endogenous interferon-β in lipopolysaccharide-triggered activation of the inducible nitric-oxide synthase gene in a mouse macrophage cell line, J774.
J Biol Chem
269
1994
12773
47
Gessani
S
Belardell
F
Pecorelli
A
Paddu
P
Baglioni
C
Bacterial lipopolysaccharide and gamma interferon induce transcription of β interferon mRNA and interferon secretion in murine macrophages.
J Virol
63
1989
2785
48
Kim
HS
Armstrong
D
Hamilton
TA
Tebo
JM
IL-10 suppresses LPS-induced KC mRNA expression via a translation-dependent decrease in mRNA stability.
J Leukoc Biol
64
1998
33
49
Zubiaga
AM
Belasco
JG
Greenberg
ME
The nonamer UUAUUUAUU is the key AU-rich sequence motif that mediates mRNA degradation.
Mol Cell Biol
15
1995
2219
50
Lagnado
CA
Brown
CY
Goodall
GJ
AUUUA is not sufficient to promote poly(A) shortening and degradation of an mRNA: The functional sequence within AU-rich elements may be UUAUUUA(U/A)(U/A).
Mol Cell Biol
14
1994
7984
51
Caput
D
Beutler
B
Hartog
K
Thayer
R
Brown-Shimer
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
52
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
53
Kontoyiannis D, Pasparakis M, Kollias G: Generation and initial characterizartion of mice lacking TNF AU rich control elements. J Leukoc Biol Suppl 16, 1996 (abstr)
54
Oquendo
P
Albertas
J
Wen
D
Graycan
JL
Derynk
R
Stiles
CD
The platelet derived growth factor-inducible KC gene encodes a secretory protein related to platelet α granules.
J Biol Chem
264
1989
4133
55
Pennica
D
Hayflick
JS
Bringman
TS
Palladino
MA
Goeddel
DV
Cloning and expression in Escherichia coli of the cDNA for tumor necrosis factor.
Proc Natl Acad Sci USA
82
1985
6060
56
Lomedico
PT
Gubler
U
Hellmann
CP
Dukovich
M
Giri
JG
Pan
YC
Collier
K
Semionow
R
Chua
AO
Mizel
SB
Cloning and expression of murine interleukin-1 cDNA in Escherichia coli.
Nature
312
1984
458
57
Gray
PW
Glaister
D
Chen
E
Goeddel
DV
Pennica
D
Two interleukin 1 genes in the mouse: Cloning and expression of the cDNA for murine interleukin 1 beta.
J Immunol
137
1986
3644
58
Sherry
B
Tekamp-Olson
P
Gallegos
C
Bauer
D
Davatelis
G
Wolpe
SD
Masiarz
F
Coit
D
Cerami
A
Resolution of the two components of macrophage inflammatory protein 1, and cloning and characterization of one of those components, macrophage inflammatory protein 1 beta.
J Exp Med
168
1988
2251
59
Tekamp-Olson
P
Gallegos
C
Bauer
D
McClain
J
Sherry
B
Fabre
M
van Deventer
S
Cerami
A
Cloning and characterization of cDNAs for murine macrophage inflammatory protein 2 and its human homologues.
J Exp Med
172
1990
911
60
Widmer
U
Yang
Z
van Deventer
S
Manogue
KR
Sherry
B
Cerami
A
Genomic structure of murine macrophage inflammatory protein-1 alpha and conservation of potential regulatory sequences with a human homolog, LD78.
J Immunol
146
1991
4031
61
Gough
NM
Metcalf
D
Gough
J
Grail
D
Dunn
AR
Structure and expression of the mRNA for murine granulocyte-macrophage colony stimulating factor.
EMBO J
4
1985
645
62
Gessani
S
Di Marzio
P
Rizza
P
Belardelli
F
Baglioni
C
Posttranscriptional regulation of interferon mRNA levels in peritoneal macrophages.
J Virol
65
1991
989
63
Peppel
K
Vinci
JM
Baglioni
C
The AU-rich sequences in the 3′ untranslated region mediate the increased turnover of interferon mRNA induced by glucocorticoids.
J Exp Med
173
1991
349
64
Peppel
K
Baglioni
C
Deadenylation and turnover of interferon-beta mRNA.
J Biol Chem
266
1991
6663
65
Weber-Nordt
RM
Meraz
MA
Schreiber
RD
Lipopolysaccharide-dependent induction of IL-10 receptor expression on murine fibroblasts.
J Immunol
153
1994
3734
66
Finbloom
DS
Winestock
KD
IL-10 induces the tyrosine phosphorylation of tyk2 and Jak1 and the differential assembly of STAT1α and STAT3 complexes in human T cells and monocytes.
J Immunol
155
1995
1079
67
Weber-Nordt
RM
Riley
JK
Greenlund
AC
Moore
KW
Darnell
JE
Schreiber
BD
Stat-3 recruitment by two distinct ligand-induced, tyrosine-phosphorylated docking sites in the interleukin-10 receptor intracellular domain.
J Biol Chem
271
1996
27954
68
O’Farrell
AM
Liu
Y
Moore
KW
Mui
ALF
IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: Evidence for Stat3-dependent and -independent pathways.
EMBO J
17
1998
1006
69
Geng
Y
Gulbins
E
Altman
A
Lotz
M
Monocyte deactivation by interleukin 10 via inhibition of tyrosine kinase activity and the Ras signaling pathway.
Proc Natl Acad Sci USA
91
1994
8602

Author notes

Address reprint requests to Thomas A. Hamilton, PhD, Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195.

Sign in via your Institution