Peripheral monocytes are short-lived and are replenished from hematopoietic stem cells whose proliferation is believed to be confined to the bone marrow. Human peripheral monocytes are assumed not to be able to proliferate. In this study we show that CD137 (ILA/4-1BB), a member of the tumor necrosis factor receptor family, induces a widespread and profound proliferation of human peripheral monocytes. Macrophage colony-stimulating factor and granulocyte-macrophage colony-stimulating factor are essential, but not sufficient for proliferation. Additional soluble autocrine factors induced by CD137 are required. Induction of proliferation is mediated via reverse signaling through a CD137 ligand, expressed constitutively by peripheral monocytes. The ability of CD137 to induce proliferation in human peripheral monocytes is not shared by any other known molecule.

CD137, ORIGINALLY NAMED 4-1BB and induced by lymphocyte activation (ILA), is a tumor necrosis factor (TNF) receptor family member and was identified in screens for receptors expressed on activated lymphocytes.1-3 CD137 is expressed by activated lymphocytes and monocytes and expression in primary cells is strictly activation dependent.4 In several nonimmune cells, expression is inducible by proinflammatory cytokines.5,6 Soluble forms of CD137 are generated by differential splicing and can be detected at enhanced concentrations in sera of patients with rheumatoid arthritis.7 The gene for human CD137 resides on chromosome 1p36, in a cluster of related genes.8 

Crosslinking of CD137 costimulates proliferation of T lymphocytes,9-11 and CD137 ligand expressed by B lymphocytes costimulates T-cell proliferation synergistically with B7.12 Injected, agonistic anti-CD137 antibodies activate T cells sufficiently for rejection of tumors and allogeneic transplants in mice.13 14 

While agonistic antibodies and the ligand to CD137 enhance lymphocyte activation, CD137 protein has the opposite effect. It inhibits proliferation of activated T lymphocytes and induces programmed cell death. These T-cell inhibitory activities of CD137 require immobilization of the protein, indicating transmission of a signal through the ligand.11 Expression of the human CD137 ligand is inducible in T lymphocytes and is constitutive in monocytes.3 

Reverse signaling through the CD137 ligand also takes place in monocytes. Immobilized recombinant CD137 protein leads to activation of the monocytes with enhanced expression of proinflammatory cytokines, inhibition of the anti-inflammatory cytokine IL-10, and induction of activation markers such as intercellular adhesion molecule-1 (ICAM).15 CD137 also prolongs monocyte survival via induction of macrophage colony-stimulating factor (M-CSF).16 

Monocytes are key regulators of immune responses, essential for the generation of beneficial antitumor and antipathogen immune reactions. Monocytes also participate in the development of harmful autoimmune diseases.17 They migrate from the circulation to sites of inflammations, attracted by signals such as chemokines, and the accumulation of monocytes/macrophages is a characteristic feature of chronic inflammation. This accumulation is further enhanced by cytokines released at inflammatory sites, such as M-CSF, granulocyte-macrophage CSF (GM-CSF), and interleukin-3 (IL-3), which prolong the survival of monocytes/macrophages.18-20 The accumulation of monocytes/macrophages at sites of inflammation has been attributed so far solely to immigration and the extension of cell survival, because human peripheral monocytes/macrophages have been considered not to be able to proliferate.19 21 

In this study we show that CD137 is able to induce a substantial degree of proliferation/endomitosis in a large proportion of human peripheral monocytes. This proliferative activity is not shared by M-CSF or any other known molecule.

Reagents.

M-CSF and neutralizing anti–M-CSF, anti–GM-CSF, and anti–IL-3 antibodies were obtained from R&D (Wiesbaden, Germany). Anti–M-CSF: clone 26730,11, protein A–purified IgG fraction of ascites fluid of murine hybridoma; anti–GM-CSF: clone 3209.1, murine monoclonal, IgG1; and anti–IL-3: protein A–purified IgG fraction of ascites fluid of murine hybridoma. Anti–HLA-DR was obtained from Becton Dickinson (Heidelberg, Germany). The antibodies were free of sodium azide. CD137-Fc protein was purchased from Alexis (Grünberg, Germany). Human IgG1 Fc protein was obtained from Accurate Chemical and Scientific Corp (Westbury, NY). TNFRII-Fc protein was a gift from Daniela Maennel (University of Regensburg).

Cells and cell culture.

Human peripheral blood mononuclear cells (PBMCs) were isolated from buffy coats of healthy volunteers. Buffy coats were diluted with 2 equal volumes of phosphate-buffered saline (PBS), overlaid onto an equal volume of Histopaque (Sigma, Deisenhofen, Germany), and spun for 20 minutes at 1,200g. PBMCs that accumulated as a white layer at the Percoll boundary were recovered and erythrocytes were lysed with 2 mL of 200 mmol/L NH4Cl, 10 mmol/L NaHCO3, and 10 mmol/L EDTA pH 7.4 for 2 minutes at room temperature. Cells were washed 2 times with PBS, pelleted at 250g, and resuspended in RPMI, 5% fetal calf serum (FCS). Primary monocytes were isolated by elutriation.22 Elutriated monocytes were more than 95% pure and contaminating T lymphocytes were less than 3% as estimated by morphology and antigenic phenotype (CD14, CD3, CD4, and CD8 expression). Cells were cultured in polystyrene dishes (Becton Dickinson, Franklin Lakes, NJ) in RPMI 1640 supplemented with 5% FCS at a concentration of 106/mL. The cell lines HL60 and MonoMac were obtained from ATCC (Manassas, VA).

Enzyme-linked immunosorbent assay (ELISA).

ELISA kits were purchased from R&D Systems (Wiesbaden, Germany) and performed according to the manufacturer’s instructions. Cytokine concentrations were determined in triplicate and are expressed as mean ± standard deviation.

Cell survival and apoptosis.

The number of living cells was determined by counting cells in 4 representative fields using an ocular with an engraved grid to ensure equal sizes of the evaluated fields. Dead and live cells were distinguished by trypan blue exclusion.

Apoptosis was determined via DNA fragmentation by the ‘Cell Death Detection ELISA Plus’ (Boehringer Mannheim, Mannheim, Germany) following instructions provided by the manufacturer. Measurements were performed in triplicate.

Cell proliferation.

For measurement of proliferation of individual cells the ‘In situ proliferation kit’ (Boehringer Mannheim) was used. A total of 3 × 105 cells were seeded per chamber of an 8-chamber slide (Falcon; Becton Dickinson, Heidelberg, Germany), coated with Fc or CD137-Fc protein and grown for 10 days. Bromodeoxyuridine (BrdU), 10 μmol/L, was added for 60 minutes. Incorporated BrdU was visualized according to kit instructions, by staining with mouse anti-BrdU and sheep anti-mouse–fluorescein isothiocyanate (FITC). Monocytes were identified by staining with a phycoerythrin-labeled, anti-CD14 antibody (2 μg/mL; Immunotech, Marseille, France). Chromatin was stained by 5 μg/mL Hoechst 33342 (Sigma, Deisenhofen, Germany) for 5 minutes.

Proliferation of cell populations was determined in 96-well microtiter plates. 105 monocytes per well were pulsed for 24 hours with 0.5 μCi 3H-thymidine, harvested, and evaluated on the TopCount microplate scintillation counter (Packard, Meriden, CT). Each condition was performed in triplicate and results are depicted as means ± standard deviation.

Statistical analysis.

Statistical significance was evaluated by the Mann Whitney U test using SPSS software (SPSS Inc, Chicago, IL).

CD137 induces apoptosis in monocytes.

CD137 has been shown to substantially prolong the survival of human primary peripheral monocytes.16 To investigate the mechanism of this activity, we tested whether CD137 prevents apoptotic cell death. Monocytes were cultured on tissue culture dishes coated with a fusion protein consisting of the extracellular domain of CD137 and the constant domain of human immunglobulin G1 (Fc). Coating was performed with a solution of 1 μg/mL protein in PBS at 4°C overnight. Untreated and Fc protein-coated plates were used as controls. Monocytes in these control groups died due to lack of stimulation and/or survival signals.23-25 In the CD137-Fc protein-coated wells, the number of living monocytes was significantly higher from day 5 of culture onward (Fig 1).

Fig. 1.

Induction of monocyte apoptosis by CD137. 105primary monocytes were cultured on immobilized Fc or CD137-Fc protein or on untreated plates (control). The extent of apoptosis (top) and the number of living cells, determined by trypan blue exclusion (bottom) were evaluated at days 1, 3, 5, and 7. Error bars indicate standard deviation. The differences in rates of apoptosis and the number of living cells between Fc and CD137-Fc treated cultures were significant starting at days 3 and 5, respectively, with P < .05. Comparable results were obtained in 3 separate experiments.

Fig. 1.

Induction of monocyte apoptosis by CD137. 105primary monocytes were cultured on immobilized Fc or CD137-Fc protein or on untreated plates (control). The extent of apoptosis (top) and the number of living cells, determined by trypan blue exclusion (bottom) were evaluated at days 1, 3, 5, and 7. Error bars indicate standard deviation. The differences in rates of apoptosis and the number of living cells between Fc and CD137-Fc treated cultures were significant starting at days 3 and 5, respectively, with P < .05. Comparable results were obtained in 3 separate experiments.

Close modal

The extent of apoptosis in the same cultures was quantified by measuring the amount of fragmented DNA (Fig 1). Quite unexpectedly, we found a higher degree of apoptosis in monocyte cultures treated with CD137 protein, compared to untreated cultures or cultures treated with the Fc control protein.

CD137 induces proliferation of peripheral monocytes.

Despite induction of apoptosis by CD137, the fact remained that CD137 prolonged the survival of the monocyte population. These seemingly contradictory results could best be reconciled by the assumption that CD137 induces proliferation of monocytes, compensating for the loss of cells through apoptosis. As measured by 3H-thymidine incorporation, CD137 induced, in fact, a strong proliferation of monocytes (Fig 2A). Proliferation correlated positively with the time monocytes were grown in the presence of CD137 protein and reached maximum levels between day 7 to 10, with a 30-fold or even higher increase in 3H-thymidine incorporation compared with control cells. There was no difference between monocytes in untreated and Fc protein-coated wells (not shown). Also, in the CD137-Fc–treated monocyte cultures, but not in control cultures, colonies or cell aggregates were observed, which could wholly or partly result from proliferating monocytes (Fig 2B).

Fig. 2.

CD137 induces proliferation of peripheral monocytes. (A) 105 monocytes were cultured on 96-well plates coated with Fc or CD137-Fc protein. Proliferation was determined daily by a 24-hour pulse with 0.5 μCi 3H-thymidine and differed significantly from day 3 onward, with P < .02. (B) Colonies/aggregates formed on CD137-treated monocytes. Photographs were taken at day 10 of culture at a magnification of 200×.

Fig. 2.

CD137 induces proliferation of peripheral monocytes. (A) 105 monocytes were cultured on 96-well plates coated with Fc or CD137-Fc protein. Proliferation was determined daily by a 24-hour pulse with 0.5 μCi 3H-thymidine and differed significantly from day 3 onward, with P < .02. (B) Colonies/aggregates formed on CD137-treated monocytes. Photographs were taken at day 10 of culture at a magnification of 200×.

Close modal

In line with the substantial increase in 3H-thymidine incorporation is that CD137-induced proliferation was rather wide-spread. Upon labeling CD137 treated monocytes with BrdU for only 1 hour, and subsequent detection with fluoresceine-labeled, anti-BrdU antibodies, 9.3% of the cells (55 ± 6 of 589 ± 43) were found to replicate their DNA (Fig 3A). Peripheral monocytes grown on Fc-coated dishes did not show incorporation of BrdU (not shown).

Fig. 3.

Verification of proliferating cells as monocytes. (A) Monocytes were grown for 8 days with immobilized CD137 protein and labeled for 60 minutes with 10 μmol/L BrdU (yellow). (B) Monocytes were cultured on immobilized CD137-Fc protein on chamber slides. After 10 days the cells were labeled for 60 minutes with 10 μmol/L BrdU (yellow). Monocytes were identified by staining for CD14 (red), and nuclei were visualized by Hoechst 33342 (blue). Similar results were obtained in 3 independent experiments for (A) and (B), respectively.

Fig. 3.

Verification of proliferating cells as monocytes. (A) Monocytes were grown for 8 days with immobilized CD137 protein and labeled for 60 minutes with 10 μmol/L BrdU (yellow). (B) Monocytes were cultured on immobilized CD137-Fc protein on chamber slides. After 10 days the cells were labeled for 60 minutes with 10 μmol/L BrdU (yellow). Monocytes were identified by staining for CD14 (red), and nuclei were visualized by Hoechst 33342 (blue). Similar results were obtained in 3 independent experiments for (A) and (B), respectively.

Close modal

We confirmed by immunocytochemistry that the proliferating cells were in fact monocytes and not other blood cells. Proliferation was determined by BrdU-incorporation and the identity of monocytes was verified by simultaneously staining for CD14, a cell surface protein specific to monocytic cells. Nuclei were visualized by staining with the DNA intercalating dye Hoechst 33342. As shown in Fig 3B, the proliferating cells have features characteristic of monocytes: they are CD14-positive, are multi-nucleated, and have the morphological appearance of monocytes/macrophages.

Other molecules that also bind to constitutively expressed molecules on monocytes, like TNFRII-Fc or anti–HLA-DR were not able to induce proliferation (Fig 4). Furthermore, CD137 had no effect on proliferation of the monocytic cell lines HL60 and MonoMac (Table 1), nor did it induce adherence and spreading of these nonadherent cells (not shown). However, the spontaneous rate of proliferation of the two lines was more than an order of magnitude higher than the rate of primary monocytes that were stimulated with CD137 for 8 days (Table1).

Fig. 4.

Specificity of CD137-induced monocyte proliferation. 105 peripheral monocytes were cultured on plates coated with 1 μg/mL of Fc, CD137-Fc protein, anti–HLA-DR antibody, or a fusion protein consisting of the extracellular domain of the type 2 TNF receptor and the constant domain of immunoglobulin G1 (TNFRII-Fc). Proliferation was determined at day 8 by 3H-thymidine incorporation and comparable results were obtained in 2 independent experiments.

Fig. 4.

Specificity of CD137-induced monocyte proliferation. 105 peripheral monocytes were cultured on plates coated with 1 μg/mL of Fc, CD137-Fc protein, anti–HLA-DR antibody, or a fusion protein consisting of the extracellular domain of the type 2 TNF receptor and the constant domain of immunoglobulin G1 (TNFRII-Fc). Proliferation was determined at day 8 by 3H-thymidine incorporation and comparable results were obtained in 2 independent experiments.

Close modal
Table 1.

Proliferative Response of Primary and Immortalized Monocytes to CD137

Uncoated Fc CD137-Fc
Primary monocytes 0.4 ± 0.2  0.5 ± 0.2  5.1 ± 1.2  
HL-60 89.7 ± 2.7  95.5 ± 4.8  92.0 ± 12.8  
MonoMac 224.0 ± 10.4  203.8 ± 28.9  225.1 ± 23.0 
Uncoated Fc CD137-Fc
Primary monocytes 0.4 ± 0.2  0.5 ± 0.2  5.1 ± 1.2  
HL-60 89.7 ± 2.7  95.5 ± 4.8  92.0 ± 12.8  
MonoMac 224.0 ± 10.4  203.8 ± 28.9  225.1 ± 23.0 

105 primary monocytes or Monomac or HL-60 cells were cultured on 96-well plates coated with Fc or CD137-Fc protein. Proliferation was determined at day 8 by a 24-hour pulse with 0.5 μCi3H-thymidine. Numbers represent means ± standard deviation of cpm × 1,000. This experiment was repeated 3 times with comparable results.

M-CSF and GM-CSF are essential but not sufficient for CD137-induced monocyte proliferation.

Because M-CSF is an essential monocyte survival factor,26,27 and because CD137 prolongs survival of monocytes via induction of M-CSF,16 it is conceivable that M-CSF would also be required for CD137-induced proliferation of monocytes. Confirming this hypothesis, neutralizing anti–M-CSF antibodies reduced CD137 induced proliferation almost completely (Fig 5A).

Fig. 5.

M-CSF and GM-CSF are essential, but not sufficient, for CD137-induced monocyte proliferation. (A) 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein. Neutralizing anti–M-CSF antibody (2 μg/mL), anti–GM-CSF antibody (2 μg/mL), and anti–IL-3 antibody (2 μg/mL) or isotype control (IgG2a; 2 μg/mL) were added where indicated. Proliferation was determined at day 10 by 3H-thymidine incorporation in triplicate conditions. (B) M-CSF and GM-CSF do not induce proliferation of monocytes. Peripheral monocytes were cultured on 96-well plates coated with Fc or CD137-Fc protein (1 μg/mL) or M-CSF and GM-CSF at indicated concentrations (ng/mL). Proliferation was determined at day 10 by 3H-thymidine incorporation. Similar results were obtained in 3 separate experiments for (A) and (B).

Fig. 5.

M-CSF and GM-CSF are essential, but not sufficient, for CD137-induced monocyte proliferation. (A) 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein. Neutralizing anti–M-CSF antibody (2 μg/mL), anti–GM-CSF antibody (2 μg/mL), and anti–IL-3 antibody (2 μg/mL) or isotype control (IgG2a; 2 μg/mL) were added where indicated. Proliferation was determined at day 10 by 3H-thymidine incorporation in triplicate conditions. (B) M-CSF and GM-CSF do not induce proliferation of monocytes. Peripheral monocytes were cultured on 96-well plates coated with Fc or CD137-Fc protein (1 μg/mL) or M-CSF and GM-CSF at indicated concentrations (ng/mL). Proliferation was determined at day 10 by 3H-thymidine incorporation. Similar results were obtained in 3 separate experiments for (A) and (B).

Close modal

Besides M-CSF, GM-CSF and IL-3 have been reported to be important for monocyte survival.18 Complete inhibition of monocyte proliferation was achieved by neutralizing M-CSF and GM-CSF. Neutralizing GM-CSF or IL-3 alone had only a slight (18% reduction) or no effect on CD137-induced proliferation, respectively. The sequestration of both GM-CSF and IL-3 together synergistically reduced proliferation to one third (Fig 5A).

These experiments established M-CSF and GM-CSF as being essential for CD137-induced proliferation of peripheral monocytes. However, they were not sufficient. M-CSF and GM-CSF at concentrations induced by CD137 elicited only a negligible proliferation (Fig 5B). M-CSF is induced by CD137 up to 10 ng/mL,16 whereas no GM-CSF could be measured (not shown). In vivo, M-CSF and GM-CSF are detectable around 10 ng/mL and 50 pg/mL, respectively.28-30 Even at significantly higher concentrations, ie, at 100 and 1 ng/mL, respectively, M-CSF and GM-CSF induced only a fraction of the proliferation observed with CD137 (Fig 5B). This may point to the existence of other, additional factors that are induced by CD137 in monocytes, and that contribute in an autocrine fashion to CD137-induced proliferation of monocytes.

CD137-induced monocyte proliferation is mediated by soluble autocrine factor(s).

Next, we wanted to evaluate whether these additional factors required for monocyte proliferation were soluble or cell-surface–bound. Supernatants of monocytes cultured for 24 hours on immobilized CD137 protein were harvested and cells were removed by centrifugation for 5 minutes at 12,000g. Transfer of this conditioned medium to untreated monocytes from the same donor dose-dependently induced cell growth (Fig 6A) and proliferation (Fig 6B). Addition of neutralizing anti–M-CSF antibodies (2 μg/mL) to conditioned medium blocked induction of cell growth and proliferation (not shown).

Fig. 6.

CD137 induces proliferation of monocytes via autocrine induction of soluble factor(s). 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein for 24 hours. 0, 10, 20, or 30 μL of conditioned supernatant of these cultures were transferred to 100 μL of new cultures with untreated monocytes. (A) After 8 days, the cells were photographed at a magnification of 300×. The bottom panel depicts monocytes grown on immobilized CD137-Fc protein for the 8 days. (B) Proliferation of the cultures from (A) was determined at day 8 by a 24-hour pulse with 0.5 μCi3H-thymidine. () Proliferation of monocytes on immobilized CD137-Fc protein. Cultures supplemented with conditioned supernatant from CD137-Fc–activated monocytes proliferated significantly stronger; P < .007. This experiment was repeated 3 times with similar results.

Fig. 6.

CD137 induces proliferation of monocytes via autocrine induction of soluble factor(s). 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein for 24 hours. 0, 10, 20, or 30 μL of conditioned supernatant of these cultures were transferred to 100 μL of new cultures with untreated monocytes. (A) After 8 days, the cells were photographed at a magnification of 300×. The bottom panel depicts monocytes grown on immobilized CD137-Fc protein for the 8 days. (B) Proliferation of the cultures from (A) was determined at day 8 by a 24-hour pulse with 0.5 μCi3H-thymidine. () Proliferation of monocytes on immobilized CD137-Fc protein. Cultures supplemented with conditioned supernatant from CD137-Fc–activated monocytes proliferated significantly stronger; P < .007. This experiment was repeated 3 times with similar results.

Close modal
Immobilization of CD137 protein is necessary for monocyte proliferation.

Induction of monocyte proliferation by CD137 required immobilization of the CD137 protein, eg, by coating of the protein to the surface of the tissue culture dishes (Fig 7). If CD137 was given as a soluble protein, after preventing its immobilization by prior coating of the dishes unspecifically with bovine serum albumin, the proliferative activity of CD137 was absent. Therefore, induction of monocyte proliferation by CD137 seems to be mediated by crosslinking of a CD137 ligand expressed by monocytes.

Fig. 7.

Immobilized, but not soluble, CD137 induces monocyte proliferation. 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein. Proliferation was determined at day 10 by 3H-thymidine incorporation. Identical results were obtained in 3 independent experiments.

Fig. 7.

Immobilized, but not soluble, CD137 induces monocyte proliferation. 105 peripheral monocytes were cultured on immobilized Fc or CD137-Fc protein. Proliferation was determined at day 10 by 3H-thymidine incorporation. Identical results were obtained in 3 independent experiments.

Close modal

Based on the survival enhancing activity of CD137,16 it was quite surprising to find a higher rate of apoptosis in CD137-treated monocytes. Considering that CD137 causes activation in monocytes,15 this situation resembles very much the phenomenon of activation induced cell death, originally described for lymphocytes. Cell activators that induce activation and proliferation simultaneously induce apoptosis in a certain percentage of the cells, but as long as proliferation outweighs cell death, the cell population expands.31 

The contradictory results of the simultaneous induction of cell survival and apoptosis led us to test whether CD137 is able to induce proliferation of monocytes. The finding that human peripheral monocytes in fact do proliferate came as a surprise. The generally held opinion is that monocytes originate from hematopoietic stem cells, the proliferation of which is confined to the bone marrow. Peripheral monocytes and macrophages are assumed not to be able to proliferate.19,21 This notion was supported by experiments measuring only marginal proliferation of peripheral monocytes, even after treatment with monocyte activators and survival factors. None or only 0.3% of peripheral monocytes were found to undergo DNA replication in response to M-CSF and GM-CSF during a 20-hour labeling period.19,25 This low percentage of proliferating cells was in line with no increase or an increase by only a factor of 2 in incorporation of 3H-thymidine induced by M-CSF.19 25 Our results confirm these earlier findings. But in contrast to M-CSF, CD137 induced a strong proliferation in a large proportion of the cells. Upon labeling for only 1 hour, 9.3% of the cells had incorporated bromodeoxyuridine into their DNA, implying that a substantial percentage of the cells were proliferating. Furthermore,3H-thymidine incorporation at its maximum was 30- to 80-fold higher in CD137-treated monocytes compared with control cells. By demonstrating that the DNA replicating cells were CD14-positive, had multiple nuclei, and the morphological appearance of monocytes, we could show unequivocally that the proliferating cells were in fact monocytes.

Further, CD137 has been shown to inhibit proliferation and to induce apoptosis in T lymphocytes.11 Therefore, these cells would not, even if present, be able to contribute to the observed proliferation.

Endomitosis and cell fusion have been shown to occur in monocytes.32,33 Proliferation of monocytes induced by CD137 likely results, to some extent, in endomitosis. Many polynucleated cells are observed in CD137-protein–treated monocyte cultures. There are two indications that they arise at least partly from endomitosis rather than solely from cell fusion: First, replicating nuclei are present in already polynucleated cells that do not show signs of cell division. Second, a decrease in the cell number, as would be expected with cell fusion, is not observed. The few small BrdU-positive cells present in CD137-treated cultures may represent monocytes that just have undergone cell division. Generally, the simultaneously ongoing processes of proliferation, endomitosis, apoptosis, and cell fusion make it impossible to quantify the number of newly emerging and dying cells. The fact that the rate of proliferation of the monocytic cell lines was more than an order of magnitude larger than that of the primary cells may be due to an in vitro selection for rapid growth and an acquisition of mutations further supporting a high proliferation rate.34 35 

For monocyte survival, M-CSF has been reported to be essential and sufficient.26 27 For proliferation of monocytes, M-CSF is also essential because sequestration of M-CSF is able to abolish proliferation almost completely. However, M-CSF is not sufficient, because at physiological and even at higher concentrations it is not able to induce proliferation of monocytes on its own. Also, the combination of M-CSF and GM-CSF failed to replicate the proliferative activity of CD137. Therefore, additional factor(s) contributing to monocyte proliferation must exist.

The ability of conditioned medium from CD137-treated monocytes to induce adherence, spreading, and proliferation in other monocytes proves (1) the soluble, and (2) the autocrine nature of these additional factor(s). Adherence and spreading can be induced by M-CSF, which is upregulated by CD137, and sequestration of M-CSF by neutralizing antibodies proves that M-CSF is also an essential factor in the conditioned medium. But M-CSF did not induce proliferation. Therefore, these additional factors may be novel cytokines or a combination of M-CSF and known cytokines.

Although significant proliferation of monocytes in isolation has not been demonstrated before, the concept that peripheral monocytes are incapable of proliferating has already been questioned by two recent studies. Local proliferation of peripheral monocytes in vivo, in a granuloma, a specialized inflammatory reaction, and in vitro, after coculturing of monocytes with aortic endothelial cells, has been reported.36,37 In both systems M-CSF was essential but not sufficient for proliferation of monocytes, a situation identical with CD137-induced monocyte proliferation. Since CD137 is expressed at sites of inflammation,4 and can be expressed by endothelial cells (H.S., unpublished observation, March 1997), CD137 could have been the factor enabling monocyte proliferation in these systems. Generally, it would be sites of inflammation, where CD137 is expressed and could induce activation and proliferation of monocytes, and thereby enhance inflammatory reactions.

It needs to be emphasized that CD137 is a cell surface receptor and the activities described here are mediated by crosslinking of a CD137 ligand and take place in the ligand-bearing monocyte. Dimerization of CD137 ligand is not sufficient to induce proliferation, because the CD137-Fc protein used in these studies dimerizes via its Fc domains and the soluble dimeric CD137 protein is not able to induce monocyte proliferation. Higher-order multimerization of the ligand—achieved by immobilization of the CD137 protein to the tissue culture plates—is required. Transduction of a signal through a CD137 ligand is also responsible for activation of monocytes and for inhibition of proliferation and induction of apoptosis in T lymphocytes.11,15 Because, in the TNF receptor and ligand families, the receptors as well as the ligands are membrane-bound molecules, a bidirectional transduction of signals is possible. Bidirectional transduction of signals through the receptor as well as the ligand has also been described for the OX40, CD40, and CD30 receptor/ligand systems, three other members of the TNF receptor and ligand families.38-41 A CD137 ligand has been isolated that is expressed constitutively by monocytes,3 thus enabling resting monocytes to respond to CD137 protein.

The proliferative activity of CD137 on peripheral monocytes is not shared by other molecules binding to cell surface proteins on monocytes, nor by well-established monocyte growth factors, like M-CSF, GM-CSF, nor any other known molecule, and constitutes an activity so far unique to CD137. Given the central position of monocytes in the regulation of immune responses, inhibition of CD137 may represent a new way to interfere with destructive autoimmune reactions. Accordingly, protective immune responses against pathogens and malignant cells may be enhanced by CD137-mediated activation of monocytic and dendritic cells, cells that are intensively studied for immune therapy for cancer.42-44 

We thank Gitte Krause for excellent technical assistance, and M. Kreutz and R. Andreesen for peripheral monocytes.

Supported by the Deutsche Forschungsgemeinschaft.

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
Kwon
BS
Weissman
SM
cDNA sequences of two inducible T-cell genes.
Proc Natl Acad Sci USA
86
1989
1963
2
Schwarz
H
Tuckwell
J
Lotz
M
A receptor induced by lymphocyte activation (ILA): A new member of the human nerve growth factor/tumor necrosis factor receptor family.
Gene
134
1993
295
3
Alderson
MR
Smith
CA
Tough
TW
Davis-Smith
T
Armitage
RJ
Falk
B
Roux
E
Baker
E
Sutherland
GR
Din
WS
Goodwin
RG
Molecular and biological characterization of human 4-1BB and its ligand.
Eur J Immunol
24
1994
2219
4
Schwarz
H
Valbracht
J
Tuckwell
J
Kempis
J
Lotz
M
ILA, the human 4-1BB homologue is inducible in lymphoid and other cell lines.
Blood
85
1995
1043
5
Setareh
M
Schwarz
H
Lotz
M
A mRNA variant encoding a soluble form of 4-1BB, a member of the murine NGF/TNF receptor family.
Gene
164
1995
311
6
von Kempis
J
Schwarz
H
Lotz
M
Differentiation-dependent and stimulus-specific expression of ILA, the human 4-1BB-homologue, in cells of mesenchymal origin.
Osteoarthritis Cartilage
5
1997
394
7
Michel
J
Langstein
J
Hofstädter
F
Schwarz
H
A soluble form of CD137 (ILA/4-1BB) is released by activated lymphocytes and is detectable in sera of patients with rheumatoid arthritis.
Eur J Immunol
28
1998
290
8
Schwarz
H
Arden
K
Lotz
M
CD137, a member of the tumor necrosis factor receptor family is located on chromosome 1p36, in a cluster of related genes, and colocalizes with several malignancies.
Biochem Biophys Res Commun
235
1997
699
9
Goodwin
RG
Din
WS
Davis-Smith
T
Anderson
DM
Gimpel
SD
Sato
TA
Maliszewski
CR
Brannan
CI
Copeland
NG
Jenkins
NA
Farrah
T
Armitage
RJ
Fanslow
WC
Smith
CA
Molecular cloning of a ligand for the inducible T cell gene 4-1BB: A member of an emerging family of cytokines with homology to tumor necrosis factor.
Eur J Immunol
10
1993
2631
10
Pollok
KE
Kim
YJ
Zhou
Z
Hurtado
J
Kim
KK
Pickard
RT
Kwon
BS
Inducible T cell antigen 4-1BB. Analysis of expression and function.
J Immunol
150
1993
771
11
Schwarz
H
Blanco
F
Valbracht
J
Kempis
J
Lotz
M
ILA, a member of the human NGF/TNF receptor family regulates T lymphocyte proliferation and survival.
Blood
87
1996
2839
12
DeBenedette
MA
Chu
NR
Pollok
KE
Hurtado
J
Wade
WF
Kwon
BS
Watts
TH
Role of 4-1BB ligand in costimulation of T lymphocyte growth and its upregulation on M12 B lymphomas by cAMP.
J Exp Med
181
1995
985
13
Melero
I
Shuford
WW
Newby
SA
Aruffo
A
Ledbetter
JA
Hellström
KE
Mittler
RS
Chen
L
Monoclonal antibodies against 4-1BB T cell activation molecule eradicate established tumors.
Nat Med
3
1997
682
14
Shuford
WW
Klussman
K
Tritchler
DD
Loo
DT
Chalupny
J
Siadak
AW
Brown
TJ
Emswiler
J
Raecho
H
Larsen
CP
Pearson
TC
Ledbetter
JA
Aruffo
A
Mittler
RS
4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the amplification in vivo of cytotoxic T cell responses.
J Exp Med
186
1997
47
15
Langstein
J
Michel
J
Fritsche
J
Kreutz
M
Andreesen
R
Schwarz
H
CD137 (ILA/4-1BB), a member of the TNF receptor family regulates monocyte activation via reverse signaling.
J Immunol
160
1998
2488
16
Langstein
J
Schwarz
H
Identification of CD137 as a potent monocyte survival factor.
J Leukoc Biol
65
1999
829
17
Lewis
CE
McGee
JO’D
The Macrophage.
1992
IRL Press at Oxford University Press
Oxford, UK
18
Young
DA
Lowe
LD
Clark
SC
Comparison of the effects of IL-3, granulocyte-macrophage colony-stimulating factor, and macrophage colony-stimulating factor in supporting monocyte differentiation in culture. Analysis of macrophage antibody-dependent cellular cytotoxicity.
J Immunol
145
1990
607
19
Xing
Z
Ohtoshi
T
Ralph
P
Gauldie
J
Jordana
M
Human upper airway structural cell-derived cytokines support human peripheral blood monocyte survival: A potential mechanism for monocyte/macrophage accumulation in the tissue.
Am J Respir Cell Mol Biol
6
1992
212
20
Bratton
DL
Hamid
Q
Boguniewicz
M
Doherty
DE
Kailey
JM
Leung
DY
Granulocyte macrophage colony-stimulating factor contributes to enhanced monocyte survival in chronic atopic dermatitis.
J Clin Invest
95
1995
211
21
van Furth
R
Raeburn
TL
van Zwet
TL
Characteristics of human mononuclear phagocytes.
Blood
54
1979
485
22
Andreesen
R
Brugger
W
Scheibenbogen
C
Kreutz
M
Leser
HG
Rehm
A
Lohr
GW
Surface phenotype analysis of human monocyte to macrophage maturation.
J Leukoc Biol
47
1990
490
23
Kreutz
M
Fritsche
J
Ackermann
U
Krause
SW
Andreesen
R
Retinoic acid inhibits monocyte to macrophage survival and differentiation.
Blood
91
1998
4796
24
Munn
DH
Beall
AC
Song
D
Wrenn
RW
Throckmorton
DC
Activation-induced apoptosis in human macrophages: Developmental regulation of a novel cell death pathway by macrophage colony-stimulating factor and interferon gamma.
J Exp Med
181
1995
127
25
Erickson-Miller
CL
Brennan
JK
Abboud
CN
Examination of survival, proliferation and cell surface antigen expression of human monocytes exposed to macrophage colony-stimulating factor (M-CSF).
Int J Cell Cloning
8
1990
346
26
Williams
GT
Smith
CA
Spooncer
E
Dexter
TM
Taylor
DR
Haemopoietic colony stimulating factors promote cell survival by suppressing apoptosis.
Nature
343
1990
76
27
Brugger
W
Kreutz
M
Andreesen
R
Macrophage colony-stimulating factor is required for human monocyte survival and acts as a cofactor for their terminal differentiation to macrophages in vitro.
J Leukoc Biol
49
1991
483
28
Kawano
Y
Takaue
Y
Minakuchi
J
Kawashima
S
Kimura
F
Motoyoshi
K
Abe
T
Sato
J
Hirao
A
Saito
S
Suzue
T
Kuroda
Y
Effects of monocyte-macrophage colony-stimulating factor (M-CSF) on in vitro erythropoiesis of marrow progenitor cells from patients with renal anemia.
Eur J Haematol
54
1995
147
29
Elner
SG
Elner
VM
Jaffe
GJ
Stuart
A
Kunkel
SL
Strieter
RM
Cytokines in proliferative diabetic retinopathy and proliferative vitreoretinopathy.
Curr Eye Res
14
1995
1045
30
Saunders
MA
Mitchell
JA
Seldon
PM
Yacoub
MH
Barnes
PJ
Giembycz
MA
Belvisi
MG
Release of granulocyte-macrophage colony stimulating factor by human cultured airway smooth muscle cells: Suppression by dexamethasone.
Br J Pharmacol
120
1997
545
31
Kabelitz
D
Pohl
T
Pechhold
K
Activation-induced cell death (apoptosis) of mature peripheral T lymphocytes.
Immunol Today
14
1993
338
32
Stahl
CP
Winton
EF
Monroe
MC
Holman
RC
Zelasky
M
Liehl
E
Myers
LA
McClure
H
Anderson
D
Evatt
BL
Recombinant human granulocyte-macrophage colony-stimulating factor promotes megakaryocyte maturation in nonhuman primates.
Exp Hematol
19
1991
810
33
Fais
S
Burgio
VL
Capobianchi
MR
Gessani
S
Pallone
F
Belardelli
F
The biological relevance of polykaryons in the immune response.
Immunol Today
18
1997
522
34
Monaco
C
Califano
D
Chiappetta
G
Mineo
A
De Franciscis
V
Vecchio
G
Santelli
G
Mutated human Kirsten ras, driven by a thyroglobulin promoter, induces a growth advantage and partially dedifferentiates rat thyroid epithelial cells in vitro.
Int J Cancer
63
1995
757
35
Leslie
NR
O’Prey
J
Bartholomew
C
Harrison
PR
An activating mutation in the kit receptor abolishes the stroma requirement for growth of ELM erythroleukemia cells, but does not prevent their differentiation in response to erythropoietin.
Blood
92
1998
4798
36
Lan
HY
Nikolic-Paterson
DJ
Mu
W
Atkins
RC
Local macrophage proliferation in multinucleated giant cell and granuloma formation in experimental Goodpasture’s syndrome.
Am J Pathol
147
1995
1214
37
Antonov
AS
Munn
DH
Kolodgie
FD
Virmani
R
Gerrity
RG
Aortic endothelial cells regulate proliferation of human monocytes in vitro via a mechanism synergistic with macrophage colony-stimulating factor Convergence at the cyclin E/p27(Kip1) regulatory checkpoint.
J Clin Invest
99
1997
2867
38
Stuber
E
Neurath
M
Calderhead
D
Fell
HP
Strober
W
Cross-linking of OX40 ligand, a member of the TNF/NGF cytokine family, induces proliferation and differentiation in murine splenic B cells.
Immunity
2
1995
507
39
Grewal
IS
Xu
J
Flavell
RA
Impairment of antigen-specific T-cell priming in mice lacking CD40 ligand.
Nature
378
1995
617
40
van Essen
D
Kikutani
H
Gray
D
CD40 ligand-transduced co-stimulation of T cells in the development of helper function.
Nature
378
1995
620
41
Wiley
RW
Goodwin
RG
Smith
CA
Reverse signaling via CD30 ligand.
J Immunol
157
1996
3635
42
Mayordomo
JI
Zorina
T
Storkus
WJ
Zitvogel
L
Garcia-Prats
MD
DeLeo
AB
Lotze
MT
Bone marrow-derived dendritic cells serve as potent adjuvants for peptide-based antitumor vaccines.
Stem Cells
15
1997
94
43
Gong
J
Chen
D
Kashiwaba
M
Kufe
D
Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells.
Nat Med
3
1997
558
44
Choudhury
A
Gajewski
JL
Liang
JC
Popat
U
Claxton
DF
Kliche
KO
Andreeff
M
Champlin
RE
Use of leukemic dendritic cells for the generation of antileukemic cellular cytotoxicity against Philadelphia chromosome-positive chronic myelogenous leukemia.
Blood
89
1997
1133

Author notes

Address reprint requests to Herbert Schwarz, Department of Pathology, University of Regensburg, 93042 Regensburg, Germany.

Sign in via your Institution