Abstract
The bone marrow microenvironment plays a decisive role in multiple myeloma progression and drug resistance. Chemokines are soluble mediators of cell migration, proliferation and survival and essentially modulate tumor progression and drug resistance. Here we investigated bone marrow-derived chemokines of naive and therapy-refractory myeloma patients and discovered that high levels of the chemokine CCL27, known so far for its role in skin inflammatory processes, correlated with worse overall survival of the patients. In addition, chemokine levels were significantly higher in samples from patients who became refractory to bortezomib at first line treatment compared to resistance at later treatment lines.
In vitro as well as in an in vivo model we could show that CCL27 triggers bortezomib-resistance of myeloma cells. This effect was strictly dependent on the expression of the respective receptor, CCR10, on stroma cells and involved the modulation of IL-10 expression, activation of myeloma survival pathways, and modulation of proteasomal activity. Drug resistance could be totally reversed by blocking CCR10 by siRNA as well as blocking IL-10 and its receptor.
From our data we suggest that blocking the CCR10/CCL27/IL-10 myeloma-stroma crosstalk is a novel therapeutic target that could be especially relevant in early refractory myeloma patients.
Keywords: myeloma, CCR10, CCL27, drug resistance, stroma cells
INTRODUCTION
Multiple myeloma is an incurable plasma cell malignancy characterized by the clonal proliferation of plasma cells in the bone marrow compartment. This specific microenvironment constitutes a sanctuary for the cancer cells supporting growth, survival and drug-resistance via cell-to-cell contact and soluble mediators (reviewed in [1]). The application of novel therapeutic agents such as proteasome inhibitors and immunomodulatory drugs, which not only target the malignant clone but also the supportive microenvironment led to improved progression-free and overall survival. Yet, myeloma patients still acquire resistance to all available therapies and finally succumb to their lethal disease.
Chemokines are a family of low molecular weight (8–10 kDa) secreted, homeostatic or proinflammatory cytokines that regulate cell trafficking via interacting with a subset of seven-transmembrane G-protein coupled receptors [2]. These chemotactic cytokines are also major players in the crosstalk between tumor cells and stroma cells and are essentially involved in metastasis, survival, development of drug resistance and escape from immune surveillance in numerous cancer entities [3–6]. Chemokines in multiple myeloma are known to contribute to the bone marrow homing of the malignant cells and ligand binding can also modulate integrin expression contributing to the development of cell adhesion-mediated drug resistance [7–9]. In addition, osteolysis, a main characteristic of advanced myeloma disease, is essentially mediated by chemokines [10]. However, a complete chemokine profile of the diseased bone marrow is still lacking and might give clues for the development of novel therapies.
CCL27, also called cutaneous T-cell attracting chemokine (C-TACK), is mostly known to cause T-cell mediated skin inflammation [11, 12] and high serum levels are of prognostic relevance in patients suffering from psoriasis and atopic dermatitis [13, 14]. The respective receptor, CCR10, is expressed on T-cells and on various subtypes of normal plasma cells. The mature B cells respond exclusively to the second ligand of the receptor, CCL28. Signaling leads to a selective accumulation of normal IgA+ plasmablasts in mucosal tissue [15–17] and of IgE+ plasmablasts in inflamed airways [18]. In addition, CCR10 ligation by CCL28 supports the maintenance of memory B-cells and contributes to survival and proliferation of long-lived IgA-producing plasma cells [19, 20]. Recently, work of Karlsson and colleagues revealed that the CCL28/CCR10 axis is an important growth and survival pathway for hematopoietic stem cells and progenitor cells in the bone marrow niche [21]. Of note, mesenchymal stem cells and bone marrow derived keratinocytes as well utilize CCR10/CCL27 signaling [22, 23].
In this study, we investigated bone marrow-derived chemokines of myeloma patients with a special focus on early and late refractory patients. We discovered that high CCL27 levels correlated with shorter overall survival of the patients and with early resistance to bortezomib treatment. We further investigated the involved mechanisms in vitro and in vivo.
RESULTS
High levels of CCL27 in the bone marrow correlate with poor overall survival and early refractory disease of myeloma patients
We screened for chemokines in bone marrow aspirates of patients and age-matched healthy donors using semi-quantitative protein arrays. Beside already known myeloma-associated chemotactic molecules, CCL27 was overexpressed in patients' samples. Concomitant Elisa analysis confirmed significant differences between myeloma (n = 45; median 4640 pg/ml; IQR 3320-7291) and healthy donor samples (n = 16; median 1620 pg/ml; IQR 947-1996; p < 0.0001, Figure 1A). Patients' data is summarized in Table 1. Utilizing cutoffs determined by receiver operating characteristics (ROC) analysis, we found that high levels of CCL27 were associated with worse overall survival of patients (Figure 1B; cutoff value = 4884 pg/ml; median survival 29 vs. 77 months, p = 0.0016). We performed multivariate analysis including CCL27 expression (high or low), sex, and stage (stage MM3B versus all other stages) as covariates. From the 45 cases, one was excluded due to missing values. Although sample numbers were low, Cox regression analysis revealed that CCL27 was an independent prognostic factor for overall survival with a hazard ratio of 4.3 [1.727 – 10.975; 95% CI, p = 0.002]. Of note, CCL27 levels did not correlate with tumor load (data not shown).
Table 1. Patients' characteristics.
Number of patients | ||
---|---|---|
age | < 65 | 23 |
> 65 | 22 | |
sex | Male | 29 |
Female | 16 | |
ISS staging | I | 6 |
II | 15 | |
III | 13 | |
Unknown | 11 | |
Durie & Salmon staging | IA | 1 |
IIIA | 31 | |
IIIB | 12 | |
Unknown | 1 | |
Immunoglobulin subtypes | IgG | 15 |
IgA | 14 | |
IgM | 1 | |
Light chains | 11 | |
Unknown | 4 |
This table displays data on patients whose naive bone marrow plasma samples were utilized for CCL27 Elisa. Data refers to the date of diagnosis.
We further analyzed untreated bone marrow plasma from patients who showed bortezomib-refractory disease at first line treatment (n = 12) compared to patients that became refractory to bortezomib at higher treatment lines (n = 18) Clinical characteristics of patients is summarized in Table 2. In a subset of first line refractory patients, CCL27 levels were significantly enhanced (Figure 1C; 1st line median 4935 pg/ml; IQR 3376-8669; other lines median 3385 pg/ml; IQR 2754-4688; p < 0.05).
Table 2. Characteristics of patients refractory to bortezomib.
Number of patients | ||
---|---|---|
age | < 65 | 13 |
> 65 | 17 | |
sex | Male | 19 |
Female | 11 | |
refractory to bortezomib | at 1st line | 12 |
2nd line and higher | 18 | |
ISS staging | I | 6 |
II | 11 | |
III | 12 | |
unknown | 1 | |
Durie&Salmon staging | IA | 5 |
IIA | 7 | |
IIIA | 12 | |
IIIB | 6 | |
Immunoglobulin subtypes | IgG | 16 |
IgA | 6 | |
Light chains only | 7 | |
Nonsecretory | 1 |
Clinical characteristics of patients who became refractory to bortezomib in the course of treatment. Data refer to date of diagnosis.
CCL27 binds uniquely to its respective receptor, CCR10, which has also a second ligand, CCL28. To gain more knowledge about possible interactions of this receptor/ligand(s) system, we measured CCL28 plasma levels as well as receptor expression on plasma cells and stroma cells. Compared to CCL27, CCL28 levels were substantially lower and even not detectable in 20/42 patients' bone marrow samples (Supplementary Figure 1A; MM median 25 pg/ml; IQR 0-245; Healthy median 349 pg/ml; IQR 282-466). We confirmed previously reported CCR10 expression on myeloma cell lines and primary plasma cells [24] in our samples and we found substantial CCR10 expression on stroma cell line HS-5 and primary fibroblasts, which we routinely use as coculture systems. Primary stroma cells isolated from the bone marrow of healthy and diseased individuals also expressed CCR10. Representative flow cytometer analyses are depicted in Figure 1D. We also detected CCL27 in the supernatants from myeloma cell lines as well as stroma cells (Supplementary Figure 1B).
Functionally, Nakayama et al. showed that CCL27 and CCL28 can induce chemotaxis of myeloma cell lines [24]. We found enhanced adhesion (Supplementary Figure 2A) and chemotaxis (Supplementary Figure 2B) in response to CCL27 for myeloma cell lines NCI-H929 and MM.1S but not for OPM-2. None of the cell lines migrated in response to the CCL28 concentrations tested and both chemokines had no impact on myeloma cell proliferation (Supplementary Figure 2C). In all cell lines, the extent of bortezomib-induced cell death was not altered by the addition of CCL27 or CCL28 (Supplementary Figure 2D). Since CCL27 is primarily known for its T cell attracting properties, we additionally investigated a possible correlation between infiltrating T cells and CCL27 levels in myeloma patients on the basis of CD62L/CD45RA expression. However, T cell subset infiltration rates did not correlate with CCL27 levels (data not shown).
In the presence of stroma cells, CCL27 rescues myeloma cells from apoptosis induced by proteasome inhibitors
We further utilized stroma-myeloma cell cocultures in order to approach the in vivo crosstalk more closely and treated the cells with different drugs. In the presence of HS-5 stroma cells, the addition of CCL27 rescued myeloma cells almost completely from bortezomib-induced cell death. Supplement of the second ligand, CCL28, had no effect (Figure 2A). Results were confirmed using primary fibroblasts (Supplementary Figure 3A). While CCL27 also blocked the induction of cell death by other proteasome inhibitors, i.e. MG-132 (Supplementary Figure 3B) and carfilzomib (Supplementary Figure 3C), efficacy of melphalan treatment was not affected (Supplementary Figure 3D). Primary stroma cells isolated from three myeloma patients also rescued myeloma cell lines (Figure 2B), and survival of CD138-sorted primary myeloma cells from four patients seeded on HS-5 layer and treated with bortezomib was ameliorated by the addition of CCL27 (Figure 2C).
CCL27 protects myeloma cells from bortezomib treatment in a xenotransplant model
Chemokines are important players throughout all species, however, caution is requested for the interpolation of experimental results between genera, especially between mouse and human (reviewed in [25]). Instead of a mouse model, we here chose a chick chorioallantoic membrane (CAM) assay modified for the use with human myeloma cells as previously reported [26, 27]. This model has also been successfully utilized to investigate tumor-stroma interactions in other human B-cell malignancies [28, 29].
Onplants containing eGFP-transfected OPM-2 myeloma cells and primary human fibroblasts in a collagen matrix were positioned onto the CAM and treated with bortezomib with/without the presence of chemokine for five days (Figure 3A, schematic view). Tumors were excised and tumor mass was quantified using an anti-GFP-Elisa. As shown, addition of CCL27 rescued myeloma cells efficiently from treatment in this setting (Figure 3A, pictures and graph). This experiment was confirmed using NCI-H929, MM.1S and untransfected OPM-2 cells (Figure 3B, pictures and graphs). In this setting, the number of proliferating myeloma cells was assessed by immunohistochemical staining with human Ki-67. Whereas CCL27 rescued myeloma cells from drug-treatment, addition of CCL28 had no impact on cell survival in both assays.
CCR10 ligation regulates stromal IL-10 levels and blocking the receptor restores drug sensitivity
We found that myeloma cells were rescued even when separated from the stroma cell layer by 0.4 μm transwells, although to a lower extent (data not shown). Therefore, we further focussed on soluble factor(s) that could be involved in this drug resistance. From almost 100 proteins tested (including cytokines, chemokines and growth factors), about 20 were expressed at significant levels. Out of them, one factor was significantly regulated in myeloma-stroma cell coculture treated with bortezomib +/− CCL27, i.e. IL-10 (Figure 4A).
IL-10 is discussed as a survival factor for myeloma cells [30] and is induced in myeloma-stroma coculture [31], which we also confirmed. Bortezomib treatment has been shown to inhibit IL-10 expression probably via blocking NF-κB signaling [32]. In our experiments utilizing HS-5 as well as primary fibroblasts (PFF), CCL27 prevented the bortezomib-induced downregulation of IL-10. The second ligand, CCL28, had no impact on IL-10 levels (Figure 4B). We also found that CCL27 induced NF-κB activity in stroma cells, and levels remained high when cells were treated with bortezomib and CCL27 (Figure 4C). However, CCL27 did not induce NF-κB activity in myeloma cell lines (data not shown). On the other hand, bortezomib treatment led to enhanced CCL27 expression in myeloma cell lines, but not in HS-5 stroma cells (Figure 4D).
Knock-down of CCR10 on HS-5 cells by siRNA resensitized cocultured myeloma cells to bortezomib (Figure 5A). Of note, downregulation of CCR10 per se significantly blocked IL-10 production in cocultures and CCL27 addition did not enhance levels (Figure 5B). Drug resistance was also reversed by the addition of neutralizing antibodies to IL-10 and IL-10 receptor (Figure 5C).
IL-10 activates survival pathways, counteracts blockade of proteasomal activity and decreases surface immunoglobulin expression of myeloma cells
In further experiments, we found that recombinant IL-10 rescued the malignant plasma cells from bortezomib-induced cell death also when cultured alone, circumventing the need for CCL27 and stromal support (Figure 6A).
IL-10 is known to signal mainly via phosphorylation of STAT-3, which we also confirmed. IL-10 induced p-p38 in MM.1S cells but not in NCI cells and had no significant impact on p-AKT and p-ERK expression in both cell lines. Bortezomib treatment did not alter phosphorylation patterns significantly at the chosen time point (Figure 6B and Supplementary Table 1). The specific rescue from bortezomib-induced cell death by CCL27 led us to further investigate the proteasomal activity of myeloma cells under treatment. Briefly, treatment-related decrease of activity was significantly restored by the addition of CCL27 and this effect was abolished by an IL-10 blocking antibody (Figure 6C). Concomitantly, surface expression of monoclonal immunoglobulin was decreased on IL-10 treated myeloma cells (NCI-H929, IgA myeloma; OPM-2, IgG myeloma) (Figure 6D), whereas we found higher levels of free immunoglobulin in the supernatants of these cells upon CCL27 +/− bortezomib treatment. This effect was blocked by the addition of an anti-IL-10 antibody (Figure 6E).
In summary, we found that CCL27, a novel chemokine in the context of the bone marrow microenvironment, confers myeloma drug resistance to proteasome inhibitors by binding to stromal CCR10 and inducing IL-10, a myeloma survival factor. Enhanced IL-10 levels foster phosphorylation of the survival molecule STAT-3 as well as modulation of proteasomal activity and shedding of immunoglobulins, contributing to myeloma survival under treatment.
DISCUSSION
Resistance to the proteasome inhibitor bortezomib, which is currently utilized in most therapeutic schemes, is an Achilles heel of myeloma therapy. About one third of myeloma patients are primary refractory to bortezomib treatment while most other patients develop resistance over time [33, 34] with no predictive markers known so far. Here, we discovered that a single chemokine, CCL27, which is upregulated in myeloma patients, induced bortezomib resistance in myeloma cells. The effect was intimately linked to stromal expression of CCR10 and the production of IL-10. Notably, blocking stromal CCR10 expression or IL-10 signalling resensitized the malignant cells to treatment.
From our data, CCL27 emerges as novel chemokine in the context of the bone marrow microenvironment. Its presence in healthy control samples suggests a homeostatic role in this compartment, which has yet to be defined. In this regard, stroma cells might be one source of this chemokine. Enhanced levels in patients correlated with lower overall survival and levels were specifically high in about 40% of primary refractory patients. We hypothesize that the significant rise in CCL27 expression found in these patients facilitates oligomerization at the receptor level leading to specific, pro-tumor signaling. Oligomerization is often required in vivo for functional output of chemokine signaling [35], which might include cytokine induction. CCL27 has been described to oligomerize over a narrow concentration range, which is known to be relatively high [36]. In line with this, we found that CCL27 levels in the range of healthy donors (up to 3000 pg/ml) could not protect myeloma cells from bortezomib-induced cell death in vitro. However, levels of the defined “high expressors” (same or higher as 5000 pg/ml) led to drug-resistance in our models. Therefore, in the patient setting, the selection of high producer myeloma cell clones and/or the observed upregulation of CCL27 in myeloma cells upon bortezomib-treatment could tip the balance in favour of the tumor. Importantly, many other cell types are involved in the microenvironmental crosstalk in myeloma (reviewed in [37]) with some of them (e.g. specific subtypes of T cells and dendritic cells) also being responsive to CCL27. Other cell types might also modulate levels of soluble CCL27 and these putative interactions could essentially contribute to the observed effect in vivo. Further in depth-investigations are certainly needed.
In any case, stromal CCR10 expression is a prerequisite for the observed rescue. Therefore, targeting this receptor could be a novel and efficient therapy for resistant and primary refractory patients. First data on the CCR10 antagonist POL7085, a protein epitope mimetic, which was recently successfully used in a model of allergic eosinophilic airway inflammation [38] might pave the way for an application in multiple myeloma. Side effects should be manageable since CCR10 knock out mice displayed mild defects of plasma cell [19] and skin-specific T cell migration [39] and showed no obvious phenotype.
Concerning the involved pathways, CCL27 is known to be activated by the NF-κB pathway [40], and should thus be down-regulated by bortezomib treatment. In our experiments, we observed upregulation of CCL27 and a CCL27-induced stabilization of IL-10 in response to bortezomib, which is totally unexpected and in contrast to current data [32, 41]. Moreover, we could show that CCL27 itself induces NF-κB signaling in stroma cells and this signaling might contribute essentially to high IL-10 levels. Since NF-κB constitutes also a major survival pathway of myeloma cells and their microenvironment in general, this counterregulation by CCL27 warrants further detailed investigations.
In our experiments, IL-10 plays an essential role in the transduction of survival. This cytokine has been shown to induce myeloma cell survival and growth and to confer sensitivity to various other gp130-activating cytokines, including IL-6 [30, 42]. Targeting IL-10 could be of additional interest in the context of tumor immune evasion, since it is known as an immunosuppressive cytokine [43]. However, in vivo, IL-10 levels are not detectable in the majority of patients with established myeloma disease and expression appears to be mostly restricted to patients suffering from plasma cell leukemia [42, 44]. Recent data utilizing an ultrasensitive Elisa approach, however, support an in vivo impact of IL-10 also in myeloma, although concentrations measured were in the low picogram range [45]. It is likely that microenvironmental IL-10 is immediately utilized by the myeloma cells and therefore efficient targeting might be hard to accomplish. In contrast, targeting chemokine/receptor crosstalk is feasible and therapeutically relevant, as has been shown e.g. for CXCL12/CXCR4 also in myeloma [46, 47].
In summary, we suggest that targeting the CCR10/CCL27 crosstalk in the myeloma microenvironment could contribute to the development of tailored therapeutic interventions bringing multiple myeloma to a chronic, manageable disease or even to cure.
MATERIALS AND METHODS
Patient samples
Plasma from bone marrow aspirates of the proximal femur during hip arthroplasty were collected through routine analysis of healthy donors (n = 16) at the Department of Orthopedic Surgery, University Hospital Innsbruck. These healthy subjects were screened for the presence of viral and bacterial infections or any bone abnormalities and did not receive immunomodulatory drugs or suffer from diseases known to influence the immune system, including autoimmune diseases or cancer. Bone marrow aspirates from the posterior iliac crest of myeloma patients (n = 45) were taken at the Department of Internal Medicine, Innsbruck Medical University. In addition, untreated samples from patients that were found to be refractory to bortezomib at first line treatment (n = 12) and at later lines (2–11; n = 18), respectively, were collected at the Department of Internal Medicine, University Hospital Brno, Czech Republic, and analyzed retrospectively. For the standardized sample preparation, heparinized bone marrow (5 ml, 10 U sodium-heparin/ml of sample) was centrifuged within 1 hour at the Tyrolean Cancer Research Insitute in Innsbruck and at Brno Hospital, respectively, at 400 g for 10 minutes and plasma was immediately stored at −80°C until further analyses. The study was conducted in accordance with the Declaration of Helsinki and ethical approval was given by the local Ethics Committee (approval numbers UN4402, UN3263, UN5064 (Innsbruck) and 16/2011 (Brno). Patients as well as healthy donors provided written informed consent. Primary myeloma cells were purified using CD138 microbeads (Miltenyi Biotec, Germany) following manufacturer's protocol and primary stroma cells were collected from bone marrow aspirates due to their adherence on plastic and flow cytometric analysis of their phenotype (CD90+/CD45−).
Cell lines, cell culture, and reagents
Human myeloma cell lines NCI-H929 and OPM-2 were purchased from DSMZ (Braunschweig, Germany), the human stroma cell line HS-5 from American Type Culture Collection (Rockville, USA), and primary foreskin fibroblasts from Promocell company (Germany). MM.1S were a generous gift from Dr. N. L. Krett [48] (Northwestern University, Chicago, USA). Cell lines were routinely grown in RPMI-1640 (Lonza, Switzerland) with 10% fetal calf serum (Sigma, USA), 100 μg/ml L-glutamine and 100 U/ml penicillin-streptomycin (Life Technologies, UK) at 37°C with 5% CO2. For MM.1S, fungizone (Fisher Scientific, Austria) was added as described [48]. Cells were obtained from DSMZ and ATCC in 2012 and expanded within 6 weeks. Frozen aliquotes were further utilized for no more than 3 months. Additionally, cell lines were routinely fingerprinted at the end of the experiments to re-confirm their origin (STR-method, Institute of Legal Medicine, Medical University of Innsbruck) and tested for mycoplasma negativity.
Other purchased reagents include Bortezomib, Carfilzomib (Sellekchem, USA), MG132 (Sigma, USA), recombinant human CCL27, CCL28 (R&D Systems, USA), IL-10 (eBioscience, USA) and the anti-human antibodies for IL-10 (clone #JES3-9D7, eBioscience, USA), IL-10 receptor (clone #3F9, Biolegend, USA), CD90 (Acris, USA), CD45, CD38, IgA, and IgG (Miltenyi Biotec, Germany), as well as CCR10 (clone #314305; R&D Systems, USA).
Protein arrays
Customized chemokine arrays (Ray Biotech, USA) were used to semiquantitatively analyze myeloma cell line supernatants and bone marrow plasma samples. Customized arrays containing antibodies against 107 chemokines, cytokines and growth factors were utilized for the analysis of coculture supernatants following exactly the manufacturer's protocol. Arrays were scanned with GenePixx 4000B microarray scanner (Molecular Devices, USA) and signal intensities at 532 nm were evaluated.
Elisa
Sandwich Elisa to measure CCL27, CCL28, IL-10, (R&D systems, Germany), immunoglobulins A and G (Cell Biolab, Inc., USA) and green fluorescent protein (GFP; eBioscience, USA), respectively, were performed following the manufacturers' protocols. The optical density was measured using benchmark microplate reader (Bio-rad laboratories, USA).
The amount of DNA-bound and thus activated NF- kB in nuclear extracts (prepared utilizing Nuclear Cell Extraction Kit, Active Motif) of HS-5 stroma cell line and myeloma cell lines NCI-H929, OPM-2, and MM.1S was quantified by Elisa using the TransAM NF-kB p65 Transcription Factor Assay kit (Active Motif North America, Carlsbad, CA) according to manufacturers' protocol and as described before [26].
Flow cytometry
Surface expression of CCR10, IgA and IgG (Miltenyi Biotec, Germany) was measured by flow cytometry. Flow cytometry was performed on a FACS Canto II flow cytometer with subsequent data analysis using FACS Diva software 7.0 (BD Biosciences, USA).
Cell adhesion assay
The adhesion of myeloma cells was performed using cellix flow system under shear stress. Vena8 biochips (Cellix Ltd, Dublin, Ireland) were coated with fibronectin+/−CCL27 and +/−CCL28, respectively, blocked with 0.2% BSA for 30 mins at room temperature and washed once with Hank's BSS medium. The cells were loaded into the coated channels at shear stress of 0.5 dynes/cm2 using Mirus nanopump (Cellix) and allowed to sit on the layer for 10 mins. Then the flow was started and continued with different shear stress of 0.5 dynes/cm2, 0.1 dynes/cm2 and 0.2 dynes/cm2 for every 30 secs. At different time points, images were recorded and analyzed by DucoCell software (Cellix).
Transwell migration assay
Myeloma cell migration was performed using an 8 μm pore sized transwell system (Corning costar, USA) coated with fibronectin (100 ng/ml). Myeloma cells at a concentration of 1 × 105 were placed on the upper chamber and the lower chamber was filled with media with or without chemokines (7.9 nM and 8.1 nM). After 8 hrs, the medium in the lower chamber along with the cells were collected and counted by FACS for 1 min. Migration rate was calculated as % increase over control.
Proliferation assay
Proliferation was measured by 3H-thymidine incorporation assays. Myeloma cells (1 × 104 cells/well) were seeded into a 96-well plate with/without the addition of chemokines as described and 1 μCi of 3H-thymidine was added to each well for the last 16 hrs of a 72 hrs incubation period. Cells were harvested onto glass fibre filters, scintillation liquid was added and radioactivity was measured using a Scintillation counter (Beckman Coulter, model LS-6500). Experiments were repeated at least 3 times in quadruplicates and statistical analyses were performed utilizing paired Student's T-test.
Apoptosis assay
Myeloma cells (1 × 105/well) were seeded in triplicates in 96-wells and incubated as indicated. In coculture experiments, myeloma cells were seeded onto 50.000 HS-5 stroma cells, i.e. in a ratio of 2:1. Survival of myeloma cells was evaluated by flow cytometry utilizing Annexin-V-APC and 7-AAD staining (BD Biosciences, USA) of gated myeloma cells (CD38high/CD45 low-neg).
Ex ovo chicken chorioallantoic membrane assay (CAM assay)
The CAM assay used in this study was essentially performed as described previously [26]. We performed two settings: first, collagen onplants containing eGFP-transfected myeloma cells and primary fibroblasts were cultured with/without the addition of CCL27, bortezomib and combinations thereof, respectively. From these xenografts, protein was prepared and tumor load was determined by anti-GFP-Elisa. Second, we seeded onplants as above utilizing myeloma cells without eGFP-transfection. For histological analysis, onplants were excised, fixed in 4% paraformaldehyde solution and further processed for paraffin sectioning and staining. Tumor growth was analyzed by staining proliferating cells with anti-human Ki-67 antibody. Tumor area was determined using the ImageJ program (NIH, USA).
Proteasome activity assay
Proteasomal activity was measured according to manufacturer's protocol using Proteasome-GloTM Chymotrypsin-Like Cell-Based Assays (Promega, USA). Luminescence was detected using a luminometer (PerkinElmer, USA) and % activity was calculated according to untreated control.
CCR10 silencing in stromal cells
HS-5 stroma cells were transfected with 25nM of a pool of 4 different siRNAs specific for CCR10 or non-target control siRNA (Dharmacon, USA) using turbofect transfection reagent (Thermo Fisher Scientific, USA) according to the manufacturer's protocol. After 24, 48 and 72 hrs, respectively, cell viability and CCR10 expression was measured by flow cytometry. Cells were seeded after 24 hrs in 96-well plates and utilized for coculture experiments.
Western Blot
One million myeloma cells were treated with/without bortezomib (5.2 nM) and IL-10 (5.5 nM) for 3 hrs. Cell lysates were examined by western blot analysis using anti-human antibodies for p-STAT-3, p-Akt, Akt, p-p38, p38, p-Erk, Erk (Cell signaling, USA), STAT-3 (Santa Cruz) and alpha-tubulin (Merck Millipore, Germany).
Statistical analyses
For all datasets displaying Gaussian distribution an unpaired t-test was used for analysis, otherwise MannWhitney U test was applied. P values < 0.05 were considered as statistically significant (*p < 0.05; **p < 0.01 and ***p < 0.001; n.s. not significant). ROC analysis was used to define cut off for high and low expression of CCL27, respectively. All analyses were performed utilizing GraphPad Prism 5 software (GraphPad Software, Inc, USA). Kaplan-Meier curves were applied for survival analysis. SPSS statistics was used for multivariate analysis.
SUPPLEMENTARY MATERIALS
Acknowledgments
We thank Univ.-Doz. Dr. Michael Fiegl and Mag. Roman Weger for participating in the retrieval of patient data.
Footnotes
CONFLICTS OF INTEREST
The authors declare that they have no competing interests.
GRANT SUPPORT
The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) OPTATIO under grant agreement n°278570, from the Österreichische Krebshilfe Tirol and from the SCRI-LIMCR GmbH.
REFERENCES
- 1.Bianchi G, Munshi NC. Pathogenesis beyond the cancer clone(s) in multiple myeloma. Blood. 2015;125:3049–3058. doi: 10.1182/blood-2014-11-568881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891–928. doi: 10.1146/annurev.immunol.22.012703.104543. [DOI] [PubMed] [Google Scholar]
- 3.Bonecchi R, Locati M, Mantovani A. Chemokines and cancer: a fatal attraction. Cancer Cell. 2011;19:434–435. doi: 10.1016/j.ccr.2011.03.017. [DOI] [PubMed] [Google Scholar]
- 4.Guo F, Wang Y, Liu J, Mok SC, Xue F, Zhang W. CXCL12/CXCR4: a symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene. 2016;35:816–826. doi: 10.1038/onc.2015.139. [DOI] [PubMed] [Google Scholar]
- 5.Yaacoub K, Pedeux R, Tarte K, Guillaudeux T. Role of the tumor microenvironment in regulating apoptosis and cancer progression. Cancer Lett. 2016 doi: 10.1016/j.canlet.2016.05.012. [DOI] [PubMed] [Google Scholar]
- 6.Zlotnik A. Chemokines and cancer. Int J Cancer. 2006;119:2026–2029. doi: 10.1002/ijc.22024. [DOI] [PubMed] [Google Scholar]
- 7.Hideshima T, Chauhan D, Hayashi T, Podar K, Akiyama M, Gupta D, Richardson P, Munshi N, Anderson KC. The biological sequelae of stromal cell-derived factor-1alpha in multiple myeloma. Mol Cancer Ther. 2002;1:539–544. [PubMed] [Google Scholar]
- 8.Johrer K, Janke K, Krugmann J, Fiegl M, Greil R. Transendothelial migration of myeloma cells is increased by tumor necrosis factor (TNF)-alpha via TNF receptor 2 and autocrine up-regulation of MCP-1. Clin Cancer Res. 2004;10:1901–1910. doi: 10.1158/1078-0432.ccr-1053-03. [DOI] [PubMed] [Google Scholar]
- 9.Vande Broek I, Asosingh K, Vanderkerken K, Straetmans N, Van Camp B, Van Riet I. Chemokine receptor CCR2 is expressed by human multiple myeloma cells and mediates migration to bone marrow stromal cell-produced monocyte chemotactic proteins MCP-1, -2 and -3. Br J Cancer. 2003;88:855–862. doi: 10.1038/sj.bjc.6600833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Abe M, Hiura K, Wilde J, Moriyama K, Hashimoto T, Ozaki S, Wakatsuki S, Kosaka M, Kido S, Inoue D, Matsumoto T. Role for macrophage inflammatory protein (MIP)-1alpha and MIP-1beta in the development of osteolytic lesions in multiple myeloma. Blood. 2002;100:2195–2202. [PubMed] [Google Scholar]
- 11.Homey B, Alenius H, Muller A, Soto H, Bowman EP, Yuan W, McEvoy L, Lauerma AI, Assmann T, Bunemann E, Lehto M, Wolff H, Yen D, et al. CCL27-CCR10 interactions regulate T cell-mediated skin inflammation. Nat Med. 2002;8:157–165. doi: 10.1038/nm0202-157. [DOI] [PubMed] [Google Scholar]
- 12.Morales J, Homey B, Vicari AP, Hudak S, Oldham E, Hedrick J, Orozco R, Copeland NG, Jenkins NA, McEvoy LM, Zlotnik A. CTACK, a skin-associated chemokine that preferentially attracts skin-homing memory T cells. Proc Natl Acad Sci USA. 1999;96:14470–14475. doi: 10.1073/pnas.96.25.14470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hijnen D, De Bruin-Weller M, Oosting B, Lebre C, De Jong E, Bruijnzeel-Koomen C, Knol E. Serum thymus and activation-regulated chemokine (TARC) and cutaneous T cell- attracting chemokine (CTACK) levels in allergic diseases: TARC and CTACK are disease-specific markers for atopic dermatitis. J Allergy Clin Immunol. 2004;113:334–340. doi: 10.1016/j.jaci.2003.12.007. [DOI] [PubMed] [Google Scholar]
- 14.Kakinuma T, Saeki H, Tsunemi Y, Fujita H, Asano N, Mitsui H, Tada Y, Wakugawa M, Watanabe T, Torii H, Komine M, Asahina A, Nakamura K, et al. Increased serum cutaneous T cell-attracting chemokine (CCL27) levels in patients with atopic dermatitis and psoriasis vulgaris. J Allergy Clin Immunol. 2003;111:592–597. doi: 10.1067/mai.2003.114. [DOI] [PubMed] [Google Scholar]
- 15.Hieshima K, Kawasaki Y, Hanamoto H, Nakayama T, Nagakubo D, Kanamaru A, Yoshie O. CC chemokine ligands 25 and 28 play essential roles in intestinal extravasation of IgA antibody-secreting cells. J Immunol. 2004;173:3668–3675. doi: 10.4049/jimmunol.173.6.3668. [DOI] [PubMed] [Google Scholar]
- 16.Lazarus NH, Kunkel EJ, Johnston B, Wilson E, Youngman KR, Butcher EC. A common mucosal chemokine (mucosae-associated epithelial chemokine/CCL28) selectively attracts IgA plasmablasts. J Immunol. 2003;170:3799–3805. doi: 10.4049/jimmunol.170.7.3799. [DOI] [PubMed] [Google Scholar]
- 17.Pan J, Kunkel EJ, Gosslar U, Lazarus N, Langdon P, Broadwell K, Vierra MA, Genovese MC, Butcher EC, Soler D. A novel chemokine ligand for CCR10 and CCR3 expressed by epithelial cells in mucosal tissues. J Immunol. 2000;165:2943–2949. doi: 10.4049/jimmunol.165.6.2943. [DOI] [PubMed] [Google Scholar]
- 18.Scanlon KM, Hawksworth RJ, Lane SJ, Mahon BP. IL-17A induces CCL28, supporting the chemotaxis of IgE-secreting B cells. Int Arch Allergy Immunol. 2011;156:51–61. doi: 10.1159/000322178. [DOI] [PubMed] [Google Scholar]
- 19.Hu S, Yang K, Yang J, Li M, Xiong N. Critical roles of chemokine receptor CCR10 in regulating memory IgA responses in intestines. Proc Natl Acad Sci USA. 2011;108:E1035–1044. doi: 10.1073/pnas.1100156108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kunkel EJ, Kim CH, Lazarus NH, Vierra MA, Soler D, Bowman EP, Butcher EC. CCR10 expression is a common feature of circulating and mucosal epithelial tissue IgA Ab-secreting cells. J Clin Invest. 2003;111:1001–1010. doi: 10.1172/JCI17244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Karlsson C, Baudet A, Miharada N, Soneji S, Gupta R, Magnusson M, Enver T, Karlsson G, Larsson J. Identification of the chemokine CCL28 as a growth and survival factor for human hematopoietic stem and progenitor cells. Blood. 2013;121:3838–3842. doi: 10.1182/blood-2013-02-481192. s3831–3815. [DOI] [PubMed] [Google Scholar]
- 22.Alexeev V, Donahue A, Uitto J, Igoucheva O. Analysis of chemotactic molecules in bone marrow-derived mesenchymal stem cells and the skin: Ccl27-Ccr10 axis as a basis for targeting to cutaneous tissues. Cytotherapy. 2013;15:171–184. doi: 10.1016/j.jcyt.2012.11.006. e171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Inokuma D, Abe R, Fujita Y, Sasaki M, Shibaki A, Nakamura H, McMillan JR, Shimizu T, Shimizu H. CTACK/CCL27 accelerates skin regeneration via accumulation of bone marrow-derived keratinocytes. Stem cells. 2006;24:2810–2816. doi: 10.1634/stemcells.2006-0264. [DOI] [PubMed] [Google Scholar]
- 24.Nakayama T, Hieshima K, Izawa D, Tatsumi Y, Kanamaru A, Yoshie O. Cutting edge: profile of chemokine receptor expression on human plasma cells accounts for their efficient recruitment to target tissues. J Immunol. 2003;170:1136–1140. doi: 10.4049/jimmunol.170.3.1136. [DOI] [PubMed] [Google Scholar]
- 25.Zlotnik A, Yoshie O, Nomiyama H. The chemokine and chemokine receptor superfamilies and their molecular evolution. Genome biology. 2006;7:243. doi: 10.1186/gb-2006-7-12-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Johrer K, Obkircher M, Neureiter D, Parteli J, Zelle-Rieser C, Maizner E, Kern J, Hermann M, Hamacher F, Merkel O, Wacht N, Zidorn C, Scheideler M, et al. Antimyeloma activity of the sesquiterpene lactone cnicin: impact on Pim-2 kinase as a novel therapeutic target. Journal of molecular medicine (Berlin, Germany) 2012;90:681–693. doi: 10.1007/s00109-011-0848-x. [DOI] [PubMed] [Google Scholar]
- 27.Martowicz A, Kern J, Gunsilius E, Untergasser G. Establishment of a human multiple myeloma xenograft model in the chicken to study tumor growth, invasion and angiogenesis. Journal of visualized experiments : JoVE. 2015:e52665. doi: 10.3791/52665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Becker J, Covelo-Fernandez A, von Bonin F, Kube D, Wilting J. Specific tumor-stroma interactions of EBV-positive Burkitt's lymphoma cells in the chick chorioallantoic membrane. Vascular cell. 2012;4:3. doi: 10.1186/2045-824X-4-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Klingenberg M, Becker J, Eberth S, Kube D, Wilting J. The chick chorioallantoic membrane as an in vivo xenograft model for Burkitt lymphoma. BMC cancer. 2014;14:339. doi: 10.1186/1471-2407-14-339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gu ZJ, Costes V, Lu ZY, Zhang XG, Pitard V, Moreau JF, Bataille R, Wijdenes J, Rossi JF, Klein B. Interleukin-10 is a growth factor for human myeloma cells by induction of an oncostatin M autocrine loop. Blood. 1996;88:3972–3986. [PubMed] [Google Scholar]
- 31.Xu S, Menu E, De Becker A, Van Camp B, Vanderkerken K, Van Riet I. Bone marrow-derived mesenchymal stromal cells are attracted by multiple myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in vitro and in vivo. Stem cells (Dayton, Ohio) 2012;30:266–279. doi: 10.1002/stem.787. [DOI] [PubMed] [Google Scholar]
- 32.Chang TP, Poltoratsky V, Vancurova I. Bortezomib inhibits expression of TGF-beta1, IL-10, and CXCR4, resulting in decreased survival and migration of cutaneous T cell lymphoma cells. Journal of immunology (Baltimore, Md : 1950) 2015;194:2942–2953. doi: 10.4049/jimmunol.1402610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kumar S, Rajkumar SV. Many facets of bortezomib resistance/susceptibility. Blood. 2008;112:2177–2178. doi: 10.1182/blood-2008-07-167767. [DOI] [PubMed] [Google Scholar]
- 34.Moreau P, Richardson PG, Cavo M, Orlowski RZ, San Miguel JF, Palumbo A, Harousseau JL. Proteasome inhibitors in multiple myeloma: 10 years later. Blood. 2012;120:947–959. doi: 10.1182/blood-2012-04-403733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Veldkamp CT, Seibert C, Peterson FC, De la Cruz NB, Haugner JC, 3rd, Basnet H, Sakmar TP, Volkman BF. Structural basis of CXCR4 sulfotyrosine recognition by the chemokine SDF-1/CXCL12. Sci Signal. 2008;1:ra4. doi: 10.1126/scisignal.1160755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Jansma AL, Kirkpatrick JP, Hsu AR, Handel TM, Nietlispach D. NMR analysis of the structure, dynamics, and unique oligomerization properties of the chemokine CCL27. J Biol Chem. 2010;285:14424–14437. doi: 10.1074/jbc.M109.091108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kawano Y, Moschetta M, Manier S, Glavey S, Gorgun GT, Roccaro AM, Anderson KC, Ghobrial IM. Targeting the bone marrow microenvironment in multiple myeloma. Immunol Rev. 2015;263:160–172. doi: 10.1111/imr.12233. [DOI] [PubMed] [Google Scholar]
- 38.Daubeuf F, Jung F, Douglas GJ, Chevalier E, Frossard N. Protective effect of a Protein Epitope Mimetic CCR10 antagonist, POL7085, in a model of allergic eosinophilic airway inflammation. Respir Res. 2015;16:77. doi: 10.1186/s12931-015-0231-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Xia M, Hu S, Fu Y, Jin W, Yi Q, Matsui Y, Yang J, McDowell MA, Sarkar S, Kalia V, Xiong N. CCR10 regulates balanced maintenance and function of resident regulatory and effector T cells to promote immune homeostasis in the skin. J Allergy Clin Immunol. 2014;134:634–644. doi: 10.1016/j.jaci.2014.03.010. e610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Riis JL, Johansen C, Vestergaard C, Otkjaer K, Kragballe K, Iversen L. CCL27 expression is regulated by both p38 MAPK and IKKbeta signalling pathways. Cytokine. 2011;56:699–707. doi: 10.1016/j.cyto.2011.09.007. [DOI] [PubMed] [Google Scholar]
- 41.Kagami S, Saeki H, Komine M, Kakinuma T, Nakamura K, Tsunemi Y, Sasaki K, Asahina A, Tamaki K. CCL28 production in HaCaT cells was mediated by different signal pathways from CCL27. Exp Dermatol. 2006;15:95–100. doi: 10.1111/j.1600-0625.2005.00390.x. [DOI] [PubMed] [Google Scholar]
- 42.Lu ZY, Zhang XG, Rodriguez C, Wijdenes J, Gu ZJ, Morel-Fournier B, Harousseau JL, Bataille R, Rossi JF, Klein B. Interleukin-10 is a proliferation factor but not a differentiation factor for human myeloma cells. Blood. 1995;85:2521–2527. [PubMed] [Google Scholar]
- 43.Mannino MH, Zhu Z, Xiao H, Bai Q, Wakefield MR, Fang Y. The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 2015 doi: 10.1016/j.canlet.2015.07.009. [DOI] [PubMed] [Google Scholar]
- 44.Stasi R, Brunetti M, Bussa S, Pagano A. Serum interleukin-10 in plasma-cell dyscrasias. Am J Hematol. 1997;54:335–337. doi: 10.1002/(sici)1096-8652(199704)54:4<335::aid-ajh15>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 45.Zingone A, Wang W, Corrigan-Cummins M, Wu SP, Plyler R, Korde N, Kwok M, Manasanch EE, Tageja N, Bhutani M, Mulquin M, Zuchlinski D, Yancey MA, et al. Altered cytokine and chemokine profiles in multiple myeloma and its precursor disease. Cytokine. 2014;69:294–297. doi: 10.1016/j.cyto.2014.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ramsey DM, McAlpine SR. Halting metastasis through CXCR4 inhibition. Bioorg Med Chem Lett. 2013;23:20–25. doi: 10.1016/j.bmcl.2012.10.138. [DOI] [PubMed] [Google Scholar]
- 47.Roccaro AM, Sacco A, Purschke WG, Moschetta M, Buchner K, Maasch C, Zboralski D, Zollner S, Vonhoff S, Mishima Y, Maiso P, Reagan MR, Lonardi S, et al. SDF-1 inhibition targets the bone marrow niche for cancer therapy. Cell Rep. 2014;9:118–128. doi: 10.1016/j.celrep.2014.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Greenstein S, Krett NL, Kurosawa Y, Ma C, Chauhan D, Hideshima T, Anderson KC, Rosen ST. Characterization of the MM. 1 human multiple myeloma (MM) cell lines: a model system to elucidate the characteristics, behavior, and signaling of steroid-sensitive and -resistant MM cells. Exp Hematol. 2003;31:271–282. doi: 10.1016/s0301-472x(03)00023-7. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.