Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage

  • Authors:
    • Hideki Matsumura
    • Tadashi Kondo
    • Koichi Ogawa
    • Takafumi Tamura
    • Kiyoshi Fukunaga
    • Soichiro Murata
    • Nobuhiro Ohkohchi
  • View Affiliations

  • Published online on: September 18, 2014     https://doi.org/10.3892/ijo.2014.2662
  • Pages: 2303-2310
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although Kupffer cells (KCs) play an important role in the liver's immune response, their role in colon cancer metastasis to the liver is unclear. We here analyzed the relationship between KCs and tumor cells (TCs) in colon cancer metastasis to the liver. Fischer 344 (F344) rats were divided into control group (KC+ group) and KC elimination group (KC‑ group), in which KC elimination was induced by Cl2MDP liposome injection. RCN‑H4 colon cancer cells were injected into the rats of both groups, and the relationship between the two types of cells was observed by intravital microscopy (IVM) for 6 h. Moreover, to investigate the effect of KCs on liver metastasis formation, KCs were eliminated at different time points before and after the TC injection. The number of metastatic nodules 2 weeks after the injection was evaluated. In the KC‑ group, IVM revealed that the number of adherent TCs had increased 1.5‑fold at 6 h after the TC injection as compared with in the KC+ group. Moreover, in the KC+ group, 74% of the TCs adhered to the KCs, and KC activation and KC phagocytosis of the TCs were observed. Two weeks after the injection, the number of metastatic nodules was significantly increased in rats in which the KCs had been eliminated before the injection, but not in rats in which the KCs had been eliminated after the injection. KC activation and KC phagocytosis of TCs decreased colon cancer cell metastasis to the liver.

Introduction

Colorectal cancer (CRC) is one of the most common cancers worldwide (1). The high morbidity and mortality associated with CRC is frequently caused by development of metastasis, i.e., liver metastasis (2). This liver metastasis determines the survival rates of CRC patients. The development of liver metastasis comprises many steps: proliferation of the primary tumor, transfer to the circulating blood, adhesion to the liver sinusoids, and extravasation and proliferation into the liver (3). Adhesion of the circulating tumor cells (TCs) to the liver sinusoids is the most important step, and thus, suppression of this TC adhesion is crucial for the control of liver metastasis. Several authors have focused on the various steps of liver metastasis in animal models, and Kupffer cells (KCs) have been revealed to play an important role in the metastasis (46). KCs had a promotive effect on liver metastasis; for example, the release of inflammatory cytokines and matrix metalloproteinase by KCs caused facilitation of adhesion, extravasation, and proliferation of TCs (711). On the other hand, a suppressive effect of KCs on liver metastasis was demonstrated by their capacity to kill TCs (1216). Therefore, the role of KCs in liver metastasis remains controversial. The previous studies analyzed data obtained from fixed specimens; i.e., they did not observe TCs in real time. However, with intravital microscopy (IVM), the interaction of KCs and TCs in the same location can be consecutively observed in real time. We sought, therefore, to analyze in real time the relationship between KCs and TCs in liver metastasis by means of IVM.

Materials and methods

Animals

Male Fischer 344 (F344) rats, weighing 200–250 g, were obtained from CLEA Japan, Inc. (Tokyo, Japan). The animal experiments were carried out in a humane manner after approval was received from the Animal Experiment Committee of the University of Tsukuba.

Cell lines

RCN-H4 cells (17,18), derived from liver metastasis of an F344 rat colon adenocarcinoma cell line, were provided by the Riken Cell Bank (Ibaraki, Japan). The cells were maintained in RPMI-1640 medium supplemented with 10% heat-inactivated FBS, 100 U/ml penicillin G, and 100 g/ml streptomycin in humidified 95% air-5% CO2 at 37°C.

KC elimination

KCs were eliminated by use of Cl2MDP liposomes, as reported by van Rooijen et al (19). The liposomes were injected into the rats via the tail vein (1 ml/rat). Twenty-four hours after the injection, no KCs were found. Repopulation of the KCs started at day 3 after the injection and was completed at day 8 (20).

IVM model

The rats were divided into two groups: the control group (KC+ group; n=6) and the KC elimination group (KC- group; n=6). In the KC+ group, calcium- and magnesium-free PBS (CMF-PBS) was injected via the tail vein (1 ml/rat) 48 h before the operation. In the KC- group, Cl2MDP liposomes were injected intravenously via the tail vein (1 ml/rat) 48 h before the operation.

Fluorescence labeling for IVM (labeling of KCs)

As previously reported, KCs were labeled with fluorescence dye by means of the liposome entrapment method (21). Fluorescently labeled phosphatidylcholine was incorporated into the liposomes according to the method of Watanabe et al (22). The fluorescent pigment used was 2-(12-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino)dodecanoyl-1-hexadecanoyl-sn-glycero-3-phosphocholine (NBD-C12-HPC) (Molecular Probes, Eugene, OR, USA). Eighty minutes before the TC injection, the liposomes (4 ml/kg) were administered via a carotid artery catheter. After that, the KCs in the rat livers were stained and were clearly delineated in the fluorescent IVM image.

Labeling of TCs

After trypsinization, the TCs were washed with CMF-PBS and kept in serum-free medium (RPMI-1640) for 60 min for reconstitution of the cell surfaces. During reconstitution, the cells were incubated with rhodamine 6G (10-7 M; Sigma-Aldrich Japan, Tokyo, Japan) to label them for the fluorescence microscopy. Subsequent to rhodamine-6G staining, the percentage of living cells exceeded 90%, as assessed by the trypan blue exclusion method. After a wash with CMF-PBS, the cells were resuspended as a single-cell solution in CMF-PBS at a final concentration of 5×106 cells/ml. No detrimental influence of the preparation on the cells’ migratory or adhesive activity was observed.

IVM

IVM were performed as previously reported (21). The rats were tracheotomized under isoflurane-induced anesthesia. After a transverse laparotomy, the left hepatic lobe was exteriorized and placed on a plate specially designed to minimize movements caused by respiration and then covered with a gridded coverslip (Olympus Corporation, Tokyo, Japan) (23). Using the gridded coverslips, selected regions of interest could be relocated at later time points. The TCs (5×106 cells) were intra-arterially injected for 60 sec (24). TC injection via portal vein caused portal tumor embolism, therefore we took a conscious dicision to TC injection via intra-artery, as reported by other authors (24,25). This route of cell application was reported not to influence the adhesive behavior of the cells within the liver sinusoids (26). The circulating TCs in the sinusoids were observed at 30 sec after the end of the TC injection. IVM was performed using a modified microscope (BX30 FLA-SP; Olympus Corporation). The hepatic microcirculation was recorded by means of a CCD camera (DVC-0; DVC, Austin, TX, USA) and a digital video recorder (GV-HD700/1; Sony, Tokyo, Japan) for offline analysis. Using objective lenses (10× 0.3 – 20× 0.7; Olympus Corporation), a final magnification of ×325–×650 was achieved on the video screen. In each rat, analysis of the liver microcirculatory para meters was performed in 10 randomly selected acini at 20, 40, 60, 120, and 360 min after the TC injection (Fig. 1). The observation time for each field was 30 sec. The grid number was recorded, so that the same acini could be observed at each time point. At the end of the observation, the rats were euthanized by exsanguination. The microcirculatory parameters were quantitatively assessed using WinROOF imaging software (version 5.0; Mitani Shoji Co., Ltd, Tokyo, Japan).

Microcirculatory analysis of KCs and TCs

The following parameters were analyzed: ) the number of adherent TCs, i.e., the TCs firmly adherent to the sinusoids for longer than 20 sec [The number of adherent TCs in the scanned acini was counted and the results were expressed as the number of adherent TCs per field (1 field = ~0.2 mm2)]; ) the number of TCs adherent to KCs, i.e., the TCs detected at the same locations as the KCs; and ) the number of KCs.

Biochemical analysis

As markers of KC activation, tumor necrosis factor (TNF)-α and interleukin (IL)-1β levels in liver tissues were measured 6 h after the TC injection using a commercial enzyme-linked immunosorbent assay kit (R&D Systems, Minneapolis, MN, USA).

Transmission electron microscopy

KCs after the TC injection were assessed by transmission electron microscopy. Two hours after the injection, the livers were quickly resected. Tissue samples from the right lobe were cut into 1-mm3 cubes and stored in 2.5% glutaraldehyde. The specimens were post-fixed with osmium tetroxide, dehydrated through a graded alcohol series, and embedded in Epon mixture. Ultrathin sections were prepared using an Ultracut S microtome (Leica Aktiengesellschaft, Vienna, Austria) and mounted on copper grids. The sections were treated with uranyl acetate and lead citrate to enhance the contrast. Specimens were examined using a Hitachi H-7000 transmission electron microscope (Hitachi, Tokyo, Japan).

KC elimination model before and after TC injection

To evaluate which timing of the KC elimination and TC injection was involved in liver metastasis, the rats were divided into five groups: the KC+ group without the Cl2MDP liposome injection (n=6); the KC- group with the Cl2MDP liposome injection 2 days before the TC injection (n=6); and the KC- group with the Cl2MDP liposome injection 1, 3, and 7 days after the TC injection (KC 1D- group, KC 3D- group, and KC 7D- group, respectively; n=6 each; Fig. 2). In all groups, 5×108 RCN-H4 cells were injected via the left carotid artery. The number of metastatic nodules in the liver was evaluated by hematoxylin and eosin (H&E) staining of the liver sections obtained 2 weeks after the TC injection.

Histologic analysis

Liver tissues were obtained from each rat, fixed with 10% formaldehyde, and embedded in paraffin. The left and medial lobes were evenly cut into 10 slices. Thin sections (4 μm) were prepared and stained with H&E. The number of the metastatic nodules was assessed using WinROOF software.

Statistical analysis

All data were expressed as the means ± standard errors of the mean (SEMs). The Mann-Whitney test and analysis of variance were used, followed by the Scheffé’s test. P<0.05 was considered significant.

Results

IVM model

The circulating TCs rapidly adhered to the sinusoids, and some adherent TCs were detected in the same locations as the KCs (Fig. 3A and B). The number of KCs in the sinusoids was significantly decreased in the KC- group when compared with the KC+ group before the TC injection (Fig. 4A and B). Cl2MDP successfully eliminated KCs from the liver tissue. No side-effects of Cl2MDP administration, such as liver injury, were observed.

Number of adherent TCs in the sinusoids

The total number of adherent TCs in the sinusoids peaked at 20 min after the TC injection and then decreased in both groups (Fig. 5A). Regardless of the presence of KCs, the total number of adherent TCs was almost the same in both groups until 2 h after the injection. However, the total number of adherent TCs at 6 h after the injection in the KC+ group was less than that in the KC- group. The adherent TCs in the same location as the KCs comprised 19% of the total number of adherent TCs at 20 min and increased over time, i.e., by 74% at 6 h (Fig. 5B).

TNF-α and IL-1β in liver tissue

The concentrations of TNF-α and IL-1β in the liver tissue were significantly elevated at 6 h after the TC injection in the KC+ group (Fig. 6A and B).

Transmission electron microscopy

In the transmission electron microscopy findings, some TCs were adherent to the KCs in the liver sinusoids. All of the TCs were phagocytosed by the KCs (Fig. 7A and B).

KC elimination model before and after TC injection

In the KC- group, the number of liver metastatic nodules was remarkably increased when compared with that in the KC+ group (Fig. 8A). A large number of metastatic nodules were observed in the portal area. The morphology of the metastatic nodules in both groups was the same (Fig. 8B). In the KC+ group, the number of metastatic nodules was 0.15±0.04 per slice for the left and middle liver lobes. In the KC- group, in which Cl2MDP liposomes had been administered 2 days before the TC injection, the metastases were markedly increased (57.55±4.41; Fig. 9). In the other three KC- groups, in which the Cl2MDP liposome injection had been administered 1–7 days after the TC injection, there were no significant differences in the number of metastases as compared with the KC+ group (Fig. 9). The numbers of metastatic nodules were 3.91±0.43, 0.19±0.04, and 0.19±0.05 in the KC 1D- group, KC 3D- group, and KC 7D- group, respectively.

Discussion

Proliferation of TCs in the liver after migration from the primary lesion causes CRC metastases to the liver. Some patients with CRC or CRC liver metastasis had TCs in the circulating blood (2730). KCs play an important role in liver metastasis; however, whether the role of KCs in the metastasis is promotive or suppressive remains unclear (616,31,32). Previous studies consisted mostly of histologic evaluations of fixed specimens (31,33,34), and the relationship between KCs and TCs in the same individual was not observed in real time. To clarify the role of KCs, especially in the early stage of liver metastasis, we used IVM to observe adhesion of TCs to the liver sinusoids. Our results showed that the total number of adherent TCs was significantly increased by elimination of KCs. Moreover, in the metastasis formation model, the number of liver metastases was increased by elimination of KCs. These results suggested that KCs suppressed liver metastasis. Furthermore, we found that the number of adherent TCs in the sinusoids in the early stage of liver metastasis was strongly related with the formation of the liver metastatic nodules thereafter.

IVM allows for consecutive observation of the same region, and we could observe the adhesion of the TCs to the sinusoids for up to 6 h after the TC injection. We found that the total number of adherent TCs in the KC+ group decreased time-dependently, and at 6 h after the TC injection, it was significantly smaller than that in the KC- group. Moreover, in the KC+ group, the percentage of adherent TCs detected at the same locations as the KCs increased time-dependently. These results strongly suggested that the KCs interacted with the TCs, and to elucidate this fact, we used transmission electron microscopy to demonstrate that the KCs phagocytosed the TCs. The result clearly indicated that the TCs adherent to the KCs were caught by the pseudopodia of the KCs and that the KCs phagocytosed the TCs, resulting in the decreased number of adherent TCs after the TC injection. Previous studies observed that a coculture of KCs and TCs led to the formation of pseudopodia in KCs and that the KCs attached to the TCs in vitro (35). Moreover, the TCs that were strongly caught by the pseudopodia of the KCs were phagocytosed and destroyed by the KCs (36). Timmers et al observed that KCs interacted with most injected TCs and phagocytosed most of them in vivo (16). Therefore, it was strongly suggested that KCs phagocytosed TCs in the early stage of liver metastasis and suppressed formation of liver metastasis.

KC phagocytosis of TCs needs activation of the KCs (36). To evaluate the activation of KCs, we investigated the cytokines involved in KCs, i.e., TNF-α and IL-1β (3739), in liver tissue. In the KC+ group, both cytokines were elevated 6 h after the TC injection. TCs have been reported to activate KCs; i.e., TCs led to secretion of TNF-α involved in KCs in vitro (33) and to increase of peroxidase activity in KCs in vivo (40). Moreover, KC activation, which was induced by OK432, an immunopotentiator prepared from streptococci, led to a decrease in liver metastasis in a rat model (6). It was postulated that this effect resulted from the augmentation of the tumoricidal activity and antigen-presenting activity of the KCs (6). Mise et al reported that OK432 increased the TNF production and tumoricidal activity of KCs in vitro (32). Therefore, the augmentation of KC tumoricidal activity might be a strategy to prevent liver metastases (6). Bayon et al demonstrated tumoricidal activity of KCs in vitro using rat colon adenocarcinoma CC531 cells, and reported that TC injection after the elimination of KCs led to increased liver metastasis in vivo study (31). From these reports and our results, we concluded that the activation of KCs was strongly associated with the TC injection.

After clarifying our results that KCs are necessary to suppress liver metastasis, we next sought to elucidate when the presence of KCs is necessary for suppression of liver metastasis. In the groups in which KCs were eliminated 1–7 days after the TC injection, the number of metastatic nodules was significantly decreased when compared with the group in which KCs were eliminated before the TC injection. It has been reported that circulating TCs were increased by surgical manipulation. Nishizaki et al reported that liver metastasis progressed after surgical manipulation in a VX-2 carcinoma cell line in a rabbit model (41). In clinical studies of patients with resectable CRC, circulating TCs were increased by surgical manipulation, and the presence of the circulating TCs negatively affected the prognosis after surgery (42). In another clinical study, circulating TCs were detected in 20–30% of patients with liver metastases from CRC before surgery, and the rate was increased to 50% during liver resection (27,43). Koch et al postulated that intraoperative tumor manipulation, which resulted in an enhanced release of TCs, caused tumor recurrence, at least in patients undergoing liver resection for colorectal metastases (27). Some studies demonstrated that the presence of circulating TCs within 24 h of CRC resection significantly increased the rate of recurrence (27,29,30). The circulating TCs, which were increased by surgical manipulation, were considered to contribute to the recurrence of liver metastasis following curative surgical resection. These findings suggested that the augmentation of KC phagocytosis of TCs had the potential to suppress liver metastasis after CRC surgical resection.

To our knowledge, this is the first study to show adhesion between KCs and TCs in real time by using IVM. The TCs adhered to the KCs, and the KCs were then activated and phagocytosed the TCs. We also demonstrated that KCs played a suppressive role in liver metastasis. In this study, the role of KCs in the early stage of liver metastasis was revealed. Future study of the detailed mechanism of KC phagocytosis of TCs may provide new therapies for the prevention of liver metastasis after curative resection of CRC.

Acknowledgements

This study was supported in part by the Japanese Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT) (Kakenhi no. 25462078). We thank F. Miyamasu for grammatical revision.

References

1 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar

2 

van der Bij GJ, Bögels M, Otten MA, et al: Experimentally induced liver metastases from colorectal cancer can be prevented by mononuclear phagocyte-mediated monoclonal antibody therapy. J Hepatol. 53:677–685. 2010.

3 

Gout S and Huot J: Role of cancer microenvironment in metastasis: focus on colon cancer. Cancer Microenviron. 1:69–83. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Williams DL, Sherwood ER, McNamee RB, Jones EL and Di Luzio NR: Therapeutic efficacy of glucan in a murine model of hepatic metastatic disease. Hepatology. 5:198–206. 1985. View Article : Google Scholar : PubMed/NCBI

5 

Heuff G, Oldenburg HS, Boutkan H, et al: Enhanced tumour growth in the rat liver after selective elimination of Kupffer cells. Cancer Immunol Immunother. 37:125–130. 1993. View Article : Google Scholar : PubMed/NCBI

6 

Zhang W, Arii S, Sasaoki T, et al: The role of Kupffer cells in the surveillance of tumor growth in the liver. J Surg Res. 55:140–146. 1993. View Article : Google Scholar : PubMed/NCBI

7 

Thomas P, Hayashi H, Zimmer R and Forse RA: Regulation of cytokine production in carcinoembryonic antigen stimulated Kupffer cells by beta-2 adrenergic receptors: implications for hepatic metastasis. Cancer Lett. 209:251–257. 2004. View Article : Google Scholar

8 

Gangopadhyay A, Lazure DA and Thomas P: Carcinoembryonic antigen induces signal transduction in Kupffer cells. Cancer Lett. 118:1–6. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Gangopadhyay A, Lazure DA and Thomas P: Adhesion of colorectal carcinoma cells to the endothelium is mediated by cytokines from CEA stimulated Kupffer cells. Clin Exp Metastasis. 16:703–712. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Auguste P, Fallavollita L, Wang N, Burnier J, Bikfalvi A and Brodt P: The host inflammatory response promotes liver metastasis by increasing tumor cell arrest and extravasation. Am J Pathol. 170:1781–1792. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Christophi C, Harun N and Fifis T: Liver regeneration and tumor stimulation - a review of cytokine and angiogenic factors. J Gastrointest Surg. 12:966–980. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Roh MS, Wang L, Oyedeji C, et al: Human Kupffer cells are cytotoxic against human colon adenocarcinoma. Surgery. 108:400–405. 1990.PubMed/NCBI

13 

Curley SA, Roh MS, Feig B, Oyedeji C, Kleinerman ES and Klostergaard J: Mechanisms of Kupffer cell cytotoxicity in vitro against the syngeneic murine colon adenocarcinoma line MCA26. J Leukoc Biol. 53:715–721. 1993.PubMed/NCBI

14 

Thomas C, Nijenhuis AM, Dontje B, Daemen T and Scherphof GL: Tumoricidal response of liver macrophages isolated from rats bearing liver metastases of colon adenocarcinoma. J Leukoc Biol. 57:617–623. 1995.PubMed/NCBI

15 

Heuff G, Steenbergen JJ, Van de Loosdrecht AA, et al: Isolation of cytotoxic Kupffer cells by a modified enzymatic assay: a methodological study. J Immunol Methods. 159:115–123. 1993. View Article : Google Scholar : PubMed/NCBI

16 

Timmers M, Vekemans K, Vermijlen D, et al: Interactions between rat colon carcinoma cells and Kupffer cells during the onset of hepatic metastasis. Int J Cancer. 112:793–802. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Inoue Y, Kashima Y, Aizawa K and Hatakeyama K: A new rat colon cancer cell line metastasizes spontaneously: biologic cha racteristics and chemotherapeutic response. Jpn J Cancer Res. 82:90–97. 1991. View Article : Google Scholar : PubMed/NCBI

18 

Okuno K, Hirai N, Lee YS, Kawai I, Shigeoka H and Yasutomi M: Involvement of liver-associated immunity in hepatic metastasis formation. J Surg Res. 75:148–152. 1998. View Article : Google Scholar : PubMed/NCBI

19 

van Rooijen N and Kors N: Effects of intracellular diphosphonates on cells of the mononuclear phagocyte system: in vivo effects of liposome-encapsulated diphosphonates on different macrophage subpopulations in the spleen. Calcif Tissue Int. 45:153–156. 1989.

20 

Bogers WM, Stad RK, Janssen DJ, et al: Kupffer cell depletion in vivo results in clearance of large-sized IgA aggregates in rats by liver endothelial cells. Clin Exp Immunol. 85:128–136. 1991. View Article : Google Scholar : PubMed/NCBI

21 

Tamura T, Kondo T, Pak S, et al: Interaction between Kupffer cells and platelets in the early period of hepatic ischemia-reperfusion injury - an in vivo study. J Surg Res. 178:443–451. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Watanabe R, Munemasa T, Matsumura M and Fujimaki M: Fluorescent liposomes for intravital staining of Kupffer cells to aid in vivo microscopy in rats. Methods Find Exp Clin Pharmacol. 29:321–327. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Kondo T, Okamoto S, Todoroki T, Hirano T, Schildberg FW and Messmer K: Application of a novel method for subsequent evaluation of sinusoids and postsinusoidal venules after ischemia-reperfusion injury of rat liver. Eur Surg Res. 30:252–258. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Enns A, Korb T, Schlüter K, et al: Alphavbeta5-integrins mediate early steps of metastasis formation. Eur J Cancer. 41:1065–1072. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Schlüter K, Gassmann P, Enns A, et al: Organ-specific metastatic tumor cell adhesion and extravasation of colon carcinoma cells with different metastatic potential. Am J Pathol. 169:1064–1073. 2006.PubMed/NCBI

26 

Haier J, Korb T, Hotz B, Spiegel HU and Senninger N: An intravital model to monitor steps of metastatic tumor cell adhesion within the hepatic microcirculation. J Gastrointest Surg. 7:507–514. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Koch M, Kienle P, Hinz U, et al: Detection of hematogenous tumor cell dissemination predicts tumor relapse in patients undergoing surgical resection of colorectal liver metastases. Ann Surg. 241:199–205. 2005. View Article : Google Scholar

28 

Yamaguchi K, Takagi Y, Aoki S, Futamura M and Saji S: Significant detection of circulating cancer cells in the blood by reverse transcriptase-polymerase chain reaction during colorectal cancer resection. Ann Surg. 232:58–65. 2000. View Article : Google Scholar

29 

Allen-Mersh TG, McCullough TK, Patel H, Wharton RQ, Glover C and Jonas SK: Role of circulating tumour cells in predicting recurrence after excision of primary colorectal carcinoma. Br J Surg. 94:96–105. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Patel H, Le Marer N, Wharton RQ, et al: Clearance of circulating tumor cells after excision of primary colorectal cancer. Ann Surg. 235:226–231. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Bayón LG, Izquierdo MA, Sirovich I, van Rooijen N, Beelen RH and Meijer S: Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver. Hepatology. 23:1224–1231. 1996.PubMed/NCBI

32 

Mise M, Arii S, Higashitsuji H, et al: Augmented local immunity in the liver by a streptococcal preparation, OK432, related to antitumor activity of hepatic macrophages. Immunopharmacology. 27:31–41. 1994. View Article : Google Scholar : PubMed/NCBI

33 

Khatib AM, Auguste P, Fallavollita L, et al: Characterization of the host proinflammatory response to tumor cells during the initial stages of liver metastasis. Am J Pathol. 167:749–759. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Higashi N, Ishii H, Fujiwara T, Morimoto-Tomita M and Irimura T: Redistribution of fibroblasts and macrophages as micrometastases develop into established liver metastases. Clin Exp Metastasis. 19:631–638. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Yonei Y, Kurose I, Fukumura D, et al: Evidence of direct interaction between Kupffer cells and colon cancer cells: an ultrastructural study of the co-culture. Liver. 14:37–44. 1994.PubMed/NCBI

36 

Gardner CR, Wasserman AJ and Laskin DL: Liver macrophage-mediated cytotoxicity toward mastocytoma cells involves phagocytosis of tumor targets. Hepatology. 14:318–324. 1991. View Article : Google Scholar : PubMed/NCBI

37 

Decker K: Biologically active products of stimulated liver macrophages (Kupffer cells). Eur J Biochem. 192:245–261. 1990. View Article : Google Scholar : PubMed/NCBI

38 

Laskin DL, Weinberger B and Laskin JD: Functional heterogeneity in liver and lung macrophages. J Leukoc Biol. 70:163–170. 2001.PubMed/NCBI

39 

Roberts RA, Ganey PE, Ju C, Kamendulis LM, Rusyn I and Klaunig JE: Role of the Kupffer cell in mediating hepatic toxicity and carcinogenesis. Toxicol Sci. 96:2–15. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Kruskal JB, Azouz A, Korideck H, et al: Hepatic colorectal cancer metastases: imaging initial steps of formation in mice. Radiology. 243:703–711. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Nishizaki T, Matsumata T, Kanematsu T, Yasunaga C and Sugimachi K: Surgical manipulation of VX2 carcinoma in the rabbit liver evokes enhancement of metastasis. J Surg Res. 49:92–97. 1990. View Article : Google Scholar : PubMed/NCBI

42 

Ito S, Nakanishi H, Hirai T, et al: Quantitative detection of CEA expressing free tumor cells in the peripheral blood of colorectal cancer patients during surgery with real-time RT-PCR on a LightCycler. Cancer Lett. 183:195–203. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Weitz J, Koch M, Kienle P, et al: Detection of hematogenic tumor cell dissemination in patients undergoing resection of liver metastases of colorectal cancer. Ann Surg. 232:66–72. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 45 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Matsumura H, Kondo T, Ogawa K, Tamura T, Fukunaga K, Murata S and Ohkohchi N: Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage. Int J Oncol 45: 2303-2310, 2014
APA
Matsumura, H., Kondo, T., Ogawa, K., Tamura, T., Fukunaga, K., Murata, S., & Ohkohchi, N. (2014). Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage. International Journal of Oncology, 45, 2303-2310. https://doi.org/10.3892/ijo.2014.2662
MLA
Matsumura, H., Kondo, T., Ogawa, K., Tamura, T., Fukunaga, K., Murata, S., Ohkohchi, N."Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage". International Journal of Oncology 45.6 (2014): 2303-2310.
Chicago
Matsumura, H., Kondo, T., Ogawa, K., Tamura, T., Fukunaga, K., Murata, S., Ohkohchi, N."Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage". International Journal of Oncology 45, no. 6 (2014): 2303-2310. https://doi.org/10.3892/ijo.2014.2662