GET THE APP

Human Mesenchymal Stem Cells Migrate toward Colon Cancer Partiall
Journal of Cell Science & Therapy

Journal of Cell Science & Therapy
Open Access

ISSN: 2157-7013

+44 1300 500008

Research Article - (2013) Volume 4, Issue 3

Human Mesenchymal Stem Cells Migrate toward Colon Cancer Partially Regulated by HMGB1

Go Hoshino1, Hiroshi Yagi1*, Hirotoshi Hasegawa1, Yoshiyuki Ishii1, Koji Okabayashi1, Hiroto Kikuchi1, Akimasa Yasuda2, Yo Mabuchi5, Masaya Nakamura2, Yumi Matsuzaki4, Hideyuki Okano3 and Yuko Kitagawa1
1Department of Surgery, School of Medicine, Keio University, Japan
2Department of Orthopedics, School of Medicine, Keio University, Japan
3Department of Physiology, School of Medicine, Keio University, Japan
4Institute of Medical Science, Tokyo Medical University, Japan
5Department of Biochemistry and Biophysics, Graduate School of Health Care, Tokyo Medical and Dental University, Japan
*Corresponding Author: Hiroshi Yagi, Department of Surgery, School of Medicine, Keio University, 35 Shinanomach, Shinjuku-ku, Tokyo, 160-8582, Japan, Tel: +81-3-3355-1211 (ext:62334), Fax: +81-3-3355-4707 Email:

Abstract

Although Mesenchymal Stem Cells (MSC) have been explored as a new clinically relevant cell type to repair injured tissue, a number of studies have highlighted the important aspect of MSC therapy. Studies have shown that systemically administered MSCs migrate to sites of malignant tumor. The focus of this study was to identify the mechanism of migration of human MSCs into cancerous tissue. First, the effect of cultured medium from cancer cell lines on modulating the migration of MSCs was evaluated, using seven different human colon cancer cell lines. Interestingly, the secretion level of High Mobility Group Box 1 (HMGB1) protein from each cell line affected the migration capacity of MSCs. In addition, recombinant human HMGB1 increased MSC’s migration capacity in a dose-dependent manner. Finally, 1×106 human MSCs were injected subcutaneously into mice (n=14) with colon cancer tumors that secreted high levels of HMGB1. Bioluminescence live image analysis showed that MSCs surrounded the tumors after injection into these mice through day 6. Immunohistochemical analysis using CD90 as a specific antibody revealed the existence of MSCs in and around the tumors as well as the secretion of local HMGB1 from the tumors detected by anti-HMGB1 antibody. These findings are critical in understanding the role of MSCs in development of solid tumors and further, they offer insight that may be useful in therapeutic application of MSCs in the treatment of malignant tumors.

Keywords: Mesenchymal stem cells; Malignant tumors; Immunohistochemical analysis; Stem cell therapy

Introduction

The migration capacity of Mesenchymal Stem Cells (MSC)s, which is considered to be a category of clinically relevant cell type, may be utilized for potential cell-based therapies. MSCs secrete an array of cytokines, chemokines, and growth factors [1], without the need for complicated culturing or handling techniques to yield clinically practical quantities of cells. The migration of MSCs into multiple inflamed or injured tissues has been studied in various models [2,3]. Indeed, isolated MSCs from bone marrow have been demonstrated to migrate toward inflamed tissues and affect via direct contact or in a paracrine fashion in the response of inflammatory cells such as macrophages [4,5], dendritic cells [6,7], and T-cells [3,8]. MSCs exist mostly in bone marrow niche [9], but also in various peripheral tissues including fat, salivary gland, tendon, skin, muscle, lung, and more recently, intestinal epithelium [10-16]. Interestingly, Da Silva Meirelles et al. and Crisan et al., suggested that perivascular lesions from a variety of tissues is a niche of MSCs, which may explain the biological features of multi-origination of MSCs [17,18]. However, the difference in the role of MSCs from these different niches such as bone marrow and perivascular lesions remains to be identified.

Recently, there has been heightened interest into the homing and migration capacity of MSCs into tumors. Since the process of tumor progression is highly related to inflammation, and since epithelialmesenchymal transition is critical in cancer development [19], the role of MSCs in carcinogenesis has emerged as a new attractive concept in cancer therapy. Although ample experimental evidence exists supporting the therapeutic potential of MSCs, the mechanism of homing and recruitment of MSCs into tumors and their potential role in malignant tissue progression is still not well understood.

MSCs have been shown to promote tumor progression by immune modulation [20]. In contrast, some studies reported that MSCs have a suppressive effect on tumor development; e.g. via modification of Akt signaling [21]. The discrepancy between these results may be due to the use of different tissue sources, individual donor variability, and injection timing of MSCs. The expression of critical receptors such as Toll-Like Receptor (TLR) is variable at various time points during the treatment [22], which may also influence on the effect of MSCs to tumor progression. Whether MSCs support or suppress tumor, it is clear that systemically administered MSCs can be recruited by and migrate toward tumors [23,24]. These findings are important as they can be used as a basis for studies to explore the utilization of engineered MSCs as novel carriers for delivery of anti-tumor agents to cancerous tissue, for the development of tumor targeted therapies.

Several different mediators have been reported to be involved in the homing process of MSCs [25-27]. Some of these molecules such as growth factors, chemokines or cytokines have already been described as mediators that can regulate cell migration toward inflammatory sites, e.g. SDF-1, IFN-γ, CCL5/CCR5, CCR2, TNF-α and some other peptides [28-32]. Among these molecules, Meng et al. investigated whether High Mobility Group Box 1 (HMGB1) protein, considered to be a proinflammatory cytokine, could enhance the biological properties of human bone marrow MSCs [33]. HMGB1 is involved in several autocrine and/or paracrine feedback mechanisms resulting in positive enforcement of its expression, and that of its receptor, TLR4, has been reported to express on MSCs [34]. Since HMGB1 stimulates the expression of molecules correlated to vascular endothelial growth factor and platelet-derived growth factor signaling by inducing their secretion from tumor cells, HMGB1 has been identified as an important modulator of tumor angiogenesis [35].

In this study, we hypothesized that HMGB1 secreted from tumors is one of the key mediators that enhances recruitment and homing capacity of MSCs. We found that several colon tumor-derived cell lines secreted different amount of HMGB1 and MSCs could respond to it in a dose-dependent manner. In addition, we demonstrated the recruitment of MSCs into colon cancer in vivo using histological analysis and live cell tracking imaging system.

Materials and Methods

MSC culture

Human-derived MSCs, which were isolated from human bone marrow (kindly provided from Dr. Yumi Matsuzaki, Tokyo Medical University), were cultured in medium composed of D-MEM supplemented with 10% Fetal Bovine Serum (FBS), 0.1mM nonessential amino acids, 1 ng/ml of basic Fibroblast Growth Factor (bFGF), 100 U/ml penicillin, 100 μg/ml streptomycin. MSCs were passaged every 5-7 days and the medium was changed on every two days. MSCs were detached by 0.05% trypsin-Ethylete Diaminetetra Acetic Acid (EDTA), and were washed with PBS and resuspended in DMEM with 2%, 5% and 10% FBS for in vitro studies or in PBS for in vivo study.

Enzyme-Linked Immunosorbent Assay (ELISA) for HMGB1

Colon-cancer cell lines (WiDr, LoVo, SW480, SW867, HT15, HCT29, HCT115) as well as Human Umbilical Vein Endothelial Cells (HUVECs) and Neonatal Human Dermal Fibroblasts (NHDF) as controls, were incubated in serum free medium overnight, and the culture supernatants were analyzed for secreted HMGB1 protein using an ELISA kit (Chondrex Inc, WA, USA) according to the manufacture’s instructions. Experiments were repeated three times.

Real time chemotaxis assay

Transwell chemotaxis assay was performed as described [36,37], with MSCs. EZ-Taxiscan chemotaxis assay was performed according to the manufacturer’s protocol (GE Healthcare, GE health care Japan, Tokyo, Japan) using HMGB1 (Shino test, Tokyo, Japan) (0, 10, 50, 100, 500 ng/ml), and a variety of supernatant of colon-cancer cell lines (WiDr, LoVo, SW480, SW867, HT15, HCT29, and HCT115) as chemoattractants. Experiments were repeated tree times.

Plasmid extraction and electroporation

Green Fluorescent Protein (GFP)-luciferase plasmid DNA (SV40- Luc) was kindly provided by Dr. Chikako Hara and Dr. Hideyuki Okano from the department of physiology, Keio University [38], and extracted from E. coli by means of NucleoBond® Xtra procedure kit (Macherey- Nagel, Düren, Germany). To transfect luciferase plasmid into MSCs with high efficiency, we used Amaxa™ Nucleofector™ technology (LONZA Japan, Tokyo, Japan) according to the manufacturer’s protocol, which provides non-viral transfection technology with high viability after transfection. 4×105 of MSCs was applied for each transfection. Before injection, transfected MSCs were cultured for 7 days and the ratio of successfully transfected cells and cell viability were evaluated by detecting GFP-luminescence using a fluorescence microscope (Olympus IX71 Tokyo Japan).

Animals and Bioluminescence Analysis

NOD-SCID mice (5 week old, 17 ± 2 g) were obtained from CLEA Japan Inc. (Tokyo, Japan). One million cells of a human colon cancer cell line (SW480), which secreted high amount of HMGB1 in in vitro experiment, were injected subcutaneously into two sites at the back of the mice. Ten days later when tumors grew to approximately 8 mm in diameter, 1×106 MSCs transfected with dual-luciferase secretory gene as described, were administrated subcutaneously into the colon-cancer bearing mice (n=14). Anti human HMGB1 antibody (Abnova, Taipei, Taiwan) was administered 300 μg/body subcutaneously as described in previous study [39] as negative control (n=4). Subsequently, bioluminescence images of the mice were taken using the in vivo imaging system (IVIS® system) (Caliper Life Science, Hopkinton, MA, USA) by exposure for one minute. The bioluminescence color images were viewed using the image software overlay. Mice were monitored at 24 hours, 48 hours, and 6 days after the injection of the cells to obtain serial fluorescence images. All experimental procedures and protocols were approved by the Animal Ethics Committee of the Keio University, School of Medicine.

Histological analysis

For histological studies, spleen, liver, lung, kidney, brain, and tumor tissues were collected at 24 hours, 48 hours, and 6 days after the injection of MSCs (n=3 for each time point). Tissues were fixed in ice-cold 4% paraformaldehyde at 4°C in PBS buffer containing 0.1% sodium azide. Paraffin-embedding was performed by dehydration of blocks in a graded series of alcohol and immersion in a mixture of 6:4 low-temperature paraffin and paraffin oil at 40°C overnight. Section with 4 μm thickness were cut, dewaxed, rehydrated, and washed in double-distilled water for 5 minutes. Sections were stained with Hematoxylin and Eosin (H&E) and visualized by microscopy. Immunofluorescence staining was performed using anti-human CD90/ Thy1 antibody [EPR3132] (Abcam Cambridge, UK) as a human MSC marker, and anti-human HMGB1 antibody (Abnova, Taipei, Taiwan). Alexa Fluor 594 goat anti-rabbit IgG (Invitrogen, Tokyo, Japan) was used as secondary antibody for rabbit anti-human CD90 antibody, and Alexa Fluor 488 goat anti-mouse IgG (Abcam, Cambridge, UK) was used as secondary antibody for mouse anti-human HMGB1 antibody. Auto-fluorescent cells including erythrocytes were distinguished from MSCs by using DAPI staining, which is a classic nuclear counterstain for immunofluorescence microscopy.

Statistic analysis

Comparisons between two groups were performed with Student’s two-tailed t-test, and more than two groups were compared by Analysis of Variance (ANOVA) and considered significance at P<0.05 as described previously [40].

Results

Human colon cancer cell lines express various amount of HMGB1

It was reported that the migration capacity of MSC toward tumors is partly regulated by HMGB1 mediated signaling [41]. We first measured secreted HMGB1 from 7 different human colon cancer cell lines (WiDr, LoVo, SW480, SW867, HT15, HCT29, and HCT115) as well as HUVEC and neonatal NHDF as controls by ELISA (Chondrex, WA, USA). HMGB1 was secreted at various concentrations from the different colon cancer cell lines (Figure 1a) as described in previous reports [42,43]. The highest secretion of HMGB1 was found in SW480 (127 ± 3.391 ng/ml), followed by LoVo (77.814 ± 9.979 ng/ml), and WiDr (68.132 ± 32.466 ng/ml), while the low level of secretion was detected from HT15 (16.409 ± 2.146 ng/ml), HT29 (45.439 ± 4.517 ng/ ml), HCT116 (50.183 ± 5.057 ng/ml), and SW867 (50.537 ± 4.26 ng/ ml) (Figure 1a). The secretion of HMGB1 from the human colon cancer cell lines was readily detectable by immunohistochemistry as shown in (Figure 1b). Based on these data, we chose SW480 as a high HMGB1- expressing cell line for the following experiments.

cell-science-therapy-colon-cancer

Figure 1: (a) The quantitative analysis of secreted HMGB1 protein from different seven colon-cancer cell line (HCT15, HT29, HCT116, SW867, WiDr, LoVo, SW480) Scale bars: 100 µm (b) Fluorescence microscope image of cultured cells expressing HMGB1 as shown in. Scale bars: 100 µm

MSCs migrate toward supernatant of colon cancer cells

An optimal chemotaxis assay system, that allowed time-lapse video monitoring of cell behavior in silicon-coated micro-channels, was used to analyze the response of MSCs to supernatant from colon cancer cell lines as a chemo-attractant. Representative images of the cells migrating in the micro-channels are shown in (Figure 2a).

MSCs moved along the concentration gradient of the supernatant, showing that they could migrate directionally toward bioactive molecules secreted by cell lines derived from colon cancer. (Figure 2b) demonstrated that the plots in the velocity-direction graph of MSCs stimulated by the supernatant from SW480 (HMGB1 high expressing cell line) tended to align to the square on the above right which showed the cells moved toward the stimulant, compared to that of HCT15 (HMGB1 low expressing cell line) which plots were in the center of the graph. The migration of MSCs was analyzed as the ratio of the cells that migrated across the micro-channel to the total number of the cells in the assay area (Figure 2c). MSCs showed significantly stronger chemotaxis toward supernatant of SW480 (63.6 ± 19.9%), which expressed higher quantities of HMGB1 compared to HCT15 (22.1 ± 21.4%) which expressed relatively lower quantities of HMGB1 (p<0.05). In addition, we compared the velocity of the migration of MSCs between different supernatant as chemo-attractants. The results showed that the velocity of the cells was higher in the micro-channels with supernatant from cell lines expressing higher HMGB1 (e.g. SW480:0.00545 ± 0.0018 μm/sec) than in those containing supernatant from cell lines with a low expression level (e.g. HCT15:0.0434 ± 0.0012 μm/sec) (p<0.05) (Figures 2d).

cell-science-therapy-microchemotaxis-chamber

Figure 2: (a) MSCs were applied to the microchemotaxis chamber to see their migration capacity. Each supernatant of different seven colon-cancer cell lines was applied to the channel. Migration capacity of MSCs to the chemotaxis was analyzed as (b) Velocity-direction plot graph (c) Proportion of migrated cells (d) The velocity of the cells. * p<0.05.

MSCs migrate in a concentration gradient of HMGB1 in a dose-dependent manner

Since resent studies indicated that MSCs could respond to HMGB1 as a chemoatractant [33], we next investigated whether the migration capacity of MSCs was dependent on the concentration of HMGB1, by using recombinant human HMGB1 in the same chemotaxis assay system. We prepared different concentrations of HMGB1 (0, 10, 50, 100, and 500 ng/ml) based on the results of ELISA assay of supernatant from the cancer cell lines as previously described. As shown in (Figure 3a), the number of cells in the channel that moved from the starting compartment increased with the incubation time and MSCs migrated further toward the opposite compartment according to the concentration of HMGB1.

cell-science-therapy-Migration-capacity

Figure 3: (a) MSCs were applied to the microchemotaxis chamber. Different concentrations of HMGB1 (0 ng/ml, 10 ng/ml, 50 ng/ml, 100 ng/ml, 500 ng/ml) were applied to the channel. Migration capacity of MSCs to the chemotaxis was analyzed as (b) Velocity-direction plot graph (c) Proportion of migrated cells (d) The velocity of the cells. * p< 0.05, N.S.: no significant

(Figure 3b) demonstrated that the plots in the velocity-direction graph of MSCs stimulated by HMGB1 in the concentration of 500 or 100 ng/ml tended to align to the square on the above right which showed the cells moved toward the stimulant, compared to that of 10 or 0 ng/ml which plots were in the center of the graph. MSCs were analyzed as the ratio of the cells that migrated across the micro-channel to the total number of the cells in the assay area (Figure 3c). The ratio of MSCs migrating toward compartments with higher concentration of HMGB1 was significantly higher (100 ng/ml: n= 57.0 ± 17.5%, 500 ng/ ml: n= 57.2 ± 16.3%) than those with lower concentrations of HMGB1 (0 ng/ml: n= 8.9 ± 7.8%, 10 ng/ml: n= 28.5 ± 20.1%, 50 ng/ml: n= 29.9 ± 17.7%) (Figure 3c). In addition, MSCs moved with higher velocity when HMGB1 was administered in certain concentrations (e.g. 50 ng/ ml: 0.00725 ± 0.0033 μm/sec, 100 ng/ml: 0.00727 ± 0.0029 μm/sec), compared to the control (0 ng/ml: 0.00585 ± 0.0035 μm/sec) (p<0.05), while the other concentrations of HMGB1 including 500ng/ml did not significantly increase their velocity compared to the control (Figure 3d).

MSCs migrate toward colon cancer expressing HMGB1

In order to assess the distribution of MSCs after the injection in human colon-cancer bearing mouse model, we used bioluminescence in vivo imaging system for cell tracking. MSCs were transfected using electroporation technique with GFP-luciferase expression vector. The viability of the cells after the transfection was 80 ± 14% (Supplementary Figure 1). First, SW480 cells (1x106) (HMGB1 high expression cancer cell line) were injected subcutaneously into both sides of the lower back of NOD-SCID mice (n=14). Ten days after the injection of the cancer cells, 1x106 MSCs were injected subcutaneously 5 to 10 mm to each tumor growth site (Figure 4a). As a negative control, 300 μg of anti-human HMGB1 antibody was administered subcutaneously into surrounding area of the tumors one day prior to the injection of MSCs. (Figure 4b) demonstrates the luciferase fluorescence of MSCs on a culture dish, detected by the same in vivo imaging system. It revealed the efficient transfection of the luciferase gene. (Figure 4c) represents the distribution of MSCs in a mouse 6 days after the injection. Luminescence signals in mice at each time point were clearly differentiated from surrounding tissues, and the higher “Tissue to Background Ratio” (TBK) value suggested that MSCs preferentially accumulated around tumor tissues and achieved their maximum value through day 6 after the injection. The signals on day 6 were localized at the peri-tumor areas and were significantly more intense than the signals on day 1, while the signals were detected almost in the same lesion at day 6 compared to day 1 when anti-HMGB1 antibody was administered (Figure 4d). Conversely, no luminescence signal could be observed in the tumor tissues on day 7 (data not shown).

cell-science-therapy-time-course

Figure 4: (a) The time course of the in vivo experiment. (b) Representative image of luciferase expression of MSCs 24 hours after the transfection. (c) In vivo fluorescence images of injected MSCs in colon-cancer mouse model at day1, day6. (d) In vivo fluorescence images of injected MSCs in colon-cancer mouse model with the administration of anti-human HMGB1 antibody at day1, day 6. (e) CD90 (MSCs) and HMGB1 was detected in the center of tumor (upper row) and peri-tumor (middle row) as arrows, while CD90 was not detected in the tumor from mice administered the antibody (lower row). Scale bars: 100 µm

Pathological evaluation of tumor and the surrounding tissue was performed (Figure 4e). The excised organ tissues including liver, lung, spleen, kidney and heart were also analyzed (Supplementary Figure 2). Histological and immunohistochemical examination of central and peripheral areas (Figure 4e; upper row and middle row) of tumors was performed by H&E and immunofluorescent staining using antihuman CD90/Thy1 antibody, and anti-human HMGB1 antibody. Human CD90 was detected in tumor tissues obtained on day 6 after the administration of MSCs both in the parenchyma and the surrounding area of the tumor where HMGB1 secretion was also obtained in the tissues; however, MSCs were rarely detected in the tumor from mice administered anti-HMGB1 antibody where the binding of the administered antibody was detected by secondary antibody (Figure 4e; lower row). In addition, MSCs were not found in the other organs, in which no signals were detected with HMGB1 antibody (Supplemental Figure 2). In addition, tumor size was evaluated through day 14 and no significant difference was detected between mice with and without the injection of MSCs (data not shown).

Discussion

The previous reports [42,44] have led to a great deal of attention in the role and function of MSCs in tumors. Among these, recent study indicated that colon cancer is one of the target tumors which can recruit MSCs by secreting bioactive molecules [45,46]. The result of this study also indicated that MSCs could respond to factors secreted from colon cancer cells and these factors increased their capacity of migration, demonstrated by the channel device experiments where cultured medium from cancer cells was used as a stimulator.

The tropism of MSCs for tumors raised wide interest regarding their potential as delivery vehicles of anti-cancer agents. Indeed, several reports described the feasibility of using these cells as anti-cancer delivery vehicles since they secrete different anti-cancer molecules such as Tissue Necrosis Factor (TNF), TNF Related Apoptosis Inducing Ligand (TRAIL) or interferon (IFN)−β by gene modulation. These studies showed sufficient effect in suppressing tumor progression [10,23,41]. However, Karnoub et al. suggested that MSCs have a supportive effect on tumor progression showing that co-injection of MSCs with breast cancer cell-line led to a higher degree of metastasis, but this effect was not significant in local tumor growth [20]. Also, the supportive effect on tumor growth by MSCs has been reported in different cancers such as colon cancer, lymphoma, and melanomas [47-49]. In contrast, several reports showed that MSCs may have a suppressive role in tumor development via p38 mitogen-activated protein kinases (MAPK) or by cell fusion [50,51]. Also, different types of tumors such as liver cancer, breast cancer, leukemia, and pancreatic cancer have been used to show their tumor suppressive effect [52-55]. On the other hand, Torsvik et al. suggested that the cross-contamination of MSCs with tumor cells could enhance their tumor supportive behavior [56]. Interestingly, Klopp et al. suggested that the increased tumor mass observed in these reports could be related to increased proliferation of MSCs in the tumor [57]. In this study, the difference in tumor growth was not observed between mice injected or not injected MSCs through day 14 after cancer cell injection. But the animal model itself could affect to these response of the tumor to MSCs as the severity of immunosuppression. Further study is needed to clarify the biological effect of MSCs on tumor progression.

The response of MSCs to different biological stimulators has been shown in in vitro studies [32,58]. However, it is controversial whether systemically administrated MSCs can home to a target tissue and migrate into it. Some reports demonstrated that MSCs migrate towards inflammatory tissue [58] as well as cancerous tissue and may potentially be used for therapeutic purposes [49,59]; however, others showed that most of the cells were trapped in lymph nodes or the lungs and disappeared soon after injection [60]. This discrepancy could be partly explained by the difference of cell source such as bone-marrow or fat tissue and whether the cells were cultured or not cultured after isolation [61]. The MSCs used in this study were isolated from human bonemarrow and cultured for least 5 to 9 passages. Thus, the results could be different from studies that used MSCs derived from other tissues, and non-cultured primary MSCs. Also the mechanism of their behavior is unclear since MSCs can secrete an array of bioactive molecules in response to the conditions in their environment [31] and change their secretome with time [62]. Among these molecules, MSCs have been reported to respond to several different stimulatory factors such as TNFα, IL-6, SDF1 or soluble TNF receptor [28,31,32,63] and to migrate faster toward inflammatory or cancerous tissues. Actually, MSCs have receptors for these stimulator molecules [64,65] and can identify the elevation of these factors and dramatically react to their presence in the environment [31]. However, the mechanism of this action is unclear.

Previous studies suggested that HMGB1 is one of the key modulators of tumor development and its receptor, the receptor for advanced glycation end products (RAGE) [55] or Toll Like Receptor4 (TLR4) [35], is expressed on variety of cancer cells [66-69] including colon cancer [69]. HMGB1 was reported to have a potential role in the regulation of cancer autophagy and apoptosis as response to the administration of anti-cancer agent [42] as well as to regulate migration and sprouting of endothelial cells as angiogenesis in tumor progression [35]. Moreover, the blockade of its receptor resulted in suppression of tumor growth and metastasis [66]. Therefore, HMGB1 has been recently identified as a potential candidate for tumor prevention therapy. In this study, we demonstrated HMGB1 was secreted in different concentrations by different cancer cell lines detected by ELISA and immunofluorescence study of cultured cells. Interestingly, MSCs have been reported to express one of these receptors TLR4 [34] and might react to HMGB1 through the TLR4 signaling pathway, which leads to the activation of NFκB that is also reported in MSCs with various stimuli [31]. Indeed, in our experiment, the chemotaxis assay using recombinant HMGB1 in different concentrations to the channel device (described in Methods) revealed that HMGB1 could modulate the migration capacity of MSCs in dose dependent manner. The result indicated that HMGB1 is one of the stimulating factors for migration of MSCs and activates their migration toward target cancer cells.

Based on these results in in vitro, we hypothesized that locally transplanted MSCs may react to the local stimulus and that their reactivity is partially linked to the activation of HMGB1 signaling pathway. To prove the concept that allo-transplanted MSCs can migrate toward solid tumors via HMGB1 secretion from the tumor, which we demonstrated by in vitro chemotaxis assay, we used a mouse model of human colon cancer and administered MSCs with or without anti- HMGB1 antibody. Indeed, the results showed that locally injected human MSCs migrated toward tumor and were both detected in the parenchyma and in the surrounding area of the tumor, while they were rarely detected in the tumor from the mice administered anti- HMGB1 antibody. These results may support the notion that MSCs in a peripheral tissue niche can respond to a tumor, which may exist a short distance from their niche, and can migrate into it. HMGB1 may regulate this process as a stimulatory molecule secreted from the tumors through TLR4 expressed on MSCs, as previously described [22]. Also our study suggested that the blockage of HMGB1 by its antibody modulated the migration capacity of locally administered MSCs in vivo. In addition, human MSCs were detected in the center and also in the edge of tumors where large numbers of HMGB1 positive cells were observed histologically. Although this is not direct proof that HMGB1 was the factor that brought about the migration of MSCs, the activation of MSCs by HMGB1 may be partially responsible for the observed migration of MSCs toward tumors. However, further study is required to clarify the effect of RAGE which is one of the key receptors of HMGB1, which is generally not presented on MSCs.

In summary, this study demonstrated that human MSCs migrate toward and into tumors in partially response to HMGB1 secreted from tumors into the milieu. These findings will help to understand the role of MSCs for tumors showing one of the key mechanisms. Since MSCs are relatively easy to obtain and grow in the laboratory, we anticipate that this study may lead to new approaches of cell therapy using MSCs to control cancer development.

Acknowledgements

This study was supported by the Sagawa Foundation for Promotion of Cancer Research to H.Y and Grant-in-Aid for Young Scientists (B) 23791497 to G.H

References

  1. González MA, Gonzalez-Rey E, Rico L, Büscher D, Delgado M (2009) Adipose-derived mesenchymal stem cells alleviate experimental colitis by inhibiting inflammatory and autoimmune responses. Gastroenterology 136: 978-989.
  2. Stappenbeck TS, Miyoshi H (2009) The role of stromal stem cells in tissue regeneration and wound repair. Science 324: 1666-1669.
  3. Ehninger A, Trumpp A (2011) The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J Exp Med 208: 421-428.
  4. Németh K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, et al. (2009) Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med 15: 42-49.
  5. Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE (2006) Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol 177: 2080-2087.
  6. Djouad F, Charbonnier LM, Bouffi C, Louis-Plence P, Bony C, et al. (2007) Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism. Stem Cells 25: 2025-2032.
  7. Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F (2005) Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 105: 2821-2827.
  8. Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, et al. (2010) Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466: 829-834.
  9. Grisendi G, Bussolari R, Cafarelli L, Petak I, Rasini V, et al. (2010) Adipose-derived mesenchymal stem cells as stable source of tumor necrosis factor-related apoptosis-inducing ligand delivery for cancer therapy. Cancer Res 70: 3718-3729.
  10. Rotter N, Oder J, Schlenke P, Lindner U, Böhrnsen F, et al. (2008) Isolation and characterization of adult stem cells from human salivary glands. Stem Cells Dev 17: 509-518.
  11. Salingcarnboriboon R, Yoshitake H, Tsuji K, Obinata M, Amagasa T, et al. (2003) Establishment of tendon-derived cell lines exhibiting pluripotent mesenchymal stem cell-like property. Exp Cell Res 287: 289-300.
  12. Toma JG, Akhavan M, Fernandes KJ, Barnabé-Heider F, Sadikot A, et al. (2001) Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol 3: 778-784.
  13. Lee JY, Qu-Petersen Z, Cao B, Kimura S, Jankowski R, et al. (2000) Clonal isolation of muscle-derived cells capable of enhancing muscle regeneration and bone healing. J Cell Biol 150: 1085-1100.
  14. Lama VN, Smith L, Badri L, Flint A, Andrei AC, et al. (2007) Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J Clin Invest 117: 989-996.
  15. Powell DW, Pinchuk IV, Saada JI, Chen X, Mifflin RC (2011) Mesenchymal cells of the intestinal lamina propria. Annu Rev Physiol 73: 213-237.
  16. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, et al. (2008) A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 3: 301-313.
  17. da Silva Meirelles L, Caplan AI, Nardi NB (2008) In search of the in vivo identity of mesenchymal stem cells. Stem Cells 26: 2287-2299.
  18. Kalluri R, Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119: 1420-1428.
  19. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, et al. (2007) Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 449: 557-563.
  20. Khakoo AY, Pati S, Anderson SA, Reid W, Elshal MF, et al. (2006) Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med 203: 1235-1247.
  21. Liotta F, Angeli R, Cosmi L, Fili L, Manuelli C, et al. (2008) Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing Notch signaling. Stem Cells 26: 279-289.
  22. Loebinger MR, Eddaoudi A, Davies D, Janes SM (2009) Mesenchymal stem cell delivery of TRAIL can eliminate metastatic cancer. Cancer Res 69: 4134-4142.
  23. Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ, et al. (2002) Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res 62: 3603-3608.
  24. van Poll D, Parekkadan B, Cho CH, Berthiaume F, Nahmias Y, et al. (2008) Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo. Hepatology 47: 1634-1643.
  25. Oh JY, Kim MK, Shin MS, Wee WR, Lee JH (2009) Cytokine secretion by human mesenchymal stem cells cocultured with damaged corneal epithelial cells. Cytokine 46: 100-103.
  26. Ren G, Zhang L, Zhao X, Xu G, Zhang Y, et al. (2008) Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell 2: 141-150.
  27. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, et al. (2004) Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 10: 858-864.
  28. Basu S, Broxmeyer HE (2009) CCR5 ligands modulate CXCL12-induced chemotaxis, adhesion, and Akt phosphorylation of human cord blood CD34+ cells. J Immunol 183: 7478-7488.
  29. Belema-Bedada F, Uchida S, Martire A, Kostin S, Braun T (2008) Efficient homing of multipotent adult mesenchymal stem cells depends on FROUNT-mediated clustering of CCR2. Cell Stem Cell 2: 566-575.
  30. Yagi H, Soto-Gutierrez A, Navarro-Alvarez N, Nahmias Y, Goldwasser Y, et al. (2010) Reactive bone marrow stromal cells attenuate systemic inflammation via sTNFR1. Mol Ther 18: 1857-1864.
  31. English K, Barry FP, Field-Corbett CP, Mahon BP (2007) IFN-gamma and TNF-alpha differentially regulate immunomodulation by murine mesenchymal stem cells. Immunol Lett 110: 91-100.
  32. Meng E, Guo Z, Wang H, Jin J, Wang J, et al. (2008) High mobility group box 1 protein inhibits the proliferation of human mesenchymal stem cells and promotes their migration and differentiation along osteoblastic pathway. Stem Cells Dev 17: 805-813.
  33. Tomchuck SL, Zwezdaryk KJ, Coffelt SB, Waterman RS, Danka ES, et al. (2008) Toll-like receptors on human mesenchymal stem cells drive their migration and immunomodulating responses. Stem Cells 26: 99-107.
  34. van Beijnum JR, Nowak-Sliwinska P, van den Boezem E, Hautvast P, Buurman WA, et al. (2013) Tumor angiogenesis is enforced by autocrine regulation of high-mobility group box 1. Oncogene 32: 363-374.
  35. Fukui Y, Hashimoto O, Sanui T, Oono T, Koga H, et al. (2001) Haematopoietic cell-specific CDM family protein DOCK2 is essential for lymphocyte migration. Nature 412: 826-831.
  36. Kunisaki Y, Nishikimi A, Tanaka Y, Takii R, Noda M, et al. (2006) DOCK2 is a Rac activator that regulates motility and polarity during neutrophil chemotaxis. J Cell Biol 174: 647-652.
  37. Hara-Miyauchi C, Tsuji O, Hanyu A, Okada S, Yasuda A, et al. (2012) Bioluminescent system for dynamic imaging of cell and animal behavior. Biochem Biophys Res Commun 419: 188-193.
  38. Ueno H, Matsuda T, Hashimoto S, Amaya F, Kitamura Y, et al. (2004) Contributions of high mobility group box protein in experimental and clinical acute lung injury. Am J Respir Crit Care Med 170: 1310-1316.
  39. Nakano K, Okada Y, Saito K, Tanikawa R, Sawamukai N, et al. (2007) Rheumatoid synovial endothelial cells produce macrophage colony-stimulating factor leading to osteoclastogenesis in rheumatoid arthritis. Rheumatology 46: 597-603.
  40. Balasubramani M, Day BW, Schoen RE, Getzenberg RH. (2006) Altered expression and localization of creatine kinase B, heterogeneous nuclear ribonucleoprotein F, and high mobility group box 1 protein in the nuclear matrix associated with colon cancer. Cancer research 66: 763-769.
  41. Tang D, Kang R, Cheh CW, Livesey KM, Liang X, et al. (2010) HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 29: 5299-5310.
  42. Dong Xda E, Ito N, Lotze MT, Demarco RA, Popovic P, et al. (2007) High mobility group box I (HMGB1) release from tumor cells after treatment: implications for development of targeted chemoimmunotherapy. J Immunother 30: 596-606.
  43. Curtin JF, Liu N, Candolfi M, Xiong W, Assi H, et al. (2009) HMGB1 mediates endogenous TLR2 activation and brain tumor regression. PLoS Med 6: e10.
  44. Kucerova L, Altanerova V, Matuskova M, Tyciakova S, Altaner C (2007) Adipose tissue-derived human mesenchymal stem cells mediated prodrug cancer gene therapy. Cancer Res 67: 6304-6313.
  45. Hung SC, Deng WP, Yang WK, Liu RS, Lee CC, et al. (2005) Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clinical cancer research: an official journal of the American Association for Cancer Research 11: 7749-7756.
  46. Ame-Thomas P, Maby-El Hajjami H, Monvoisin C, Jean R, Monnier D, et al. (2007) Human mesenchymal stem cells isolated from bone marrow and lymphoid organs support tumor B-cell growth: role of stromal cells in follicular lymphoma pathogenesis. Blood 109: 693-702.
  47. Djouad F, Plence P, Bony C, Tropel P, Apparailly F, et al. (2003) Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 102: 3837-3844.
  48. Shinagawa K, Kitadai Y, Tanaka M, Sumida T, Kodama M, et al. (2010) Mesenchymal stem cells enhance growth and metastasis of colon cancer. Int J Cancer 127: 2323-2333.
  49. Tian K, Yang S, Ren Q, Han Z, Lu S, et al. (2010) p38 MAPK contributes to the growth inhibition of leukemic tumor cells mediated by human umbilical cord mesenchymal stem cells. Cell Physiol Biochem 26: 799-808.
  50. Wang Y, Fan H, Zhou B, Ju Z, Yu L, et al. (2012) Fusion of human umbilical cord mesenchymal stem cells with esophageal carcinoma cells inhibits the tumorigenicity of esophageal carcinoma cells. Int J Oncol 40: 370-377.
  51. Qiao L, Xu ZL, Zhao TJ, Ye LH, Zhang XD (2008) Dkk-1 secreted by mesenchymal stem cells inhibits growth of breast cancer cells via depression of Wnt signalling. Cancer Lett 269: 67-77.
  52. Zhu Y, Sun Z, Han Q, Liao L, Wang J, et al. (2009) Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia 23: 925-933.
  53. Qiao L, Xu Z, Zhao T, Zhao Z, Shi M, et al. (2008) Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res 18: 500-507.
  54. Cousin B, Ravet E, Poglio S, De Toni F, Bertuzzi M, et al. (2009) Adult stromal cells derived from human adipose tissue provoke pancreatic cancer cell death both in vitro and in vivo. PLoS One 4: e6278.
  55. Torsvik A, Rosland GV, Svendsen A, Molven A, Immervoll H, et al. (2010) Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: putting the research field on track - letter. Cancer Res 70: 6393-6396.
  56. Klopp AH, Gupta A, Spaeth E, Andreeff M, Marini F 3rd (2011) Concise review: Dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth? Stem Cells 29: 11-19.
  57. Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, et al. (2007) The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem Cells 25: 1737-1745.
  58. Sumida T, Kitadai Y, Shinagawa K, Tanaka M, Kodama M, et al. (2011) Anti-stromal therapy with imatinib inhibits growth and metastasis of gastric carcinoma in an orthotopic nude mouse model. Int J Cancer 128: 2050-2062.
  59. Morikawa S, Mabuchi Y, Kubota Y, Nagai Y, Niibe K, et al. (2009) Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med 206: 2483-2496.
  60. Tang D, Lotze MT, Kang R, Zeh HJ (2011) Apoptosis promotes early tumorigenesis. Oncogene 30: 1851-1854.
  61. Crisostomo PR, Wang M, Herring CM, Markel TA, Meldrum KK, et al. (2007) Gender differences in injury induced mesenchymal stem cell apoptosis and VEGF, TNF, IL-6 expression: role of the 55 kDa TNF receptor (TNFR1). J Mol Cell Cardiol 42: 142-149.
  62. Markel TA, Crisostomo PR, Wang M, Herring CM, Meldrum DR. (2007) Activation of individual tumor necrosis factor receptors differentially affects stem cell growth factor and cytokine production. Am J Physiol Gastrointest Liver Physiol 293: G657-662.
  63. Ip JE, Wu Y, Huang J, Zhang L, Pratt RE, et al. (2007) Mesenchymal stem cells use integrin beta1 not CXC chemokine receptor 4 for myocardial migration and engraftment. Mol Biol Cell 18: 2873-2882.
  64. Taguchi A, Blood DC, del Toro G, Canet A, Lee DC, et al. (2000) Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature 405: 354-360.
  65. Chicoine MR, Zahner M, Won EK, Kalra RR, Kitamura T, et al. (2007) The in vivo antitumoral effects of lipopolysaccharide against glioblastoma multiforme are mediated in part by Toll-like receptor 4. Neurosurgery 60: 372-380.
  66. Garay RP, Viens P, Bauer J, Normier G, Bardou M, et al. (2007) Cancer relapse under chemotherapy: why TLR2/4 receptor agonists can help. Eur J Pharmacol 563: 1-17.
  67. Núñez NG, Andreani V, Crespo MI, Nocera DA, Breser ML, et al. (2012) IFNβ produced by TLR4-activated tumor cells is involved in improving the antitumoral immune response. Cancer Res 72: 592-603.
  68. Sasahira T, Akama Y, Fujii K, Kuniyasu H (2005) Expression of receptor for advanced glycation end products and HMGB1/amphoterin in colorectal adenomas. Virchows Arch 446: 411-415.
Citation: Hoshino G, Yagi H, Hasegawa H, Ishii Y, Okabayashi K, et al. (2013) Human Mesenchymal Stem Cells Migrate toward Colon Cancer Partially Regulated by HMGB1. J Cell Sci Ther 4:145.

Copyright: © 2013 Hoshino G, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top