Open Access

Intramuscular injection of adenoviral hepatocyte growth factor at a distal site ameliorates dextran sodium sulfate-induced colitis in mice

  • Authors:
    • Kentaro Yuge
    • Tomoyuki Takahashi
    • Ngin Cin Khai
    • Kazuko Goto
    • Takako Fujiwara
    • Hisayoshi Fujiwara
    • Ken-Ichiro Kosai
  • View Affiliations

  • Published online on: March 6, 2014     https://doi.org/10.3892/ijmm.2014.1686
  • Pages: 1064-1074
  • Copyright: © Yuge et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Inflammatory bowel disease (IBD) severely affects the quality of life of patients. At present, there is no clinical solution for this condition; therefore, there is a need for innovative therapies for IBD. Hepatocyte growth factor (HGF) exerts various biological activities in various organs. However, a clinically applicable and effective HGF-based therapy for IBD has yet to be developed. In this study, we examined the therapeutic effect of injecting an adenoviral vector encoding the human HGF gene (Ad.HGF) into the hindlimbs of mice with dextran sodium sulfate (DSS)-induced colitis. Plasma levels of circulating human HGF (hHGF) were measured in injected mice. The results showed that weight loss and colon shortening were significantly lower in Ad.HGF-infected mice as compared to control (Ad.LacZ-infected) colitic mice. Additionally, inflammation and crypt scores were significantly reduced in the entire length of the colon, particularly in the distal section. This therapeutic effect was associated with increased cell proliferation and an antiapoptotic effect, as well as a reduction in the number of CD4+ cells and a decreased CD4/CD8 ratio. The levels of inflammatory, as well as Th1 and Th2 cytokines were higher in Ad.HGF-infected mice as compared to the control colitic mice. Thus, systemically circulating hHGF protein, produced by an adenovirally transduced hHGF gene introduced at distal sites in the limbs, significantly ameliorated DSS-induced colitis by promoting cell proliferation (i.e., regeneration), preventing apoptosis, and immunomodulation. Owing to its clinical feasibility and potent therapeutic effects, this method may be developed into a clinical therapy for treating IBD.

Introduction

The breakdown of normal mucosal immunity causes the development of inflammatory bowel disease (IBD), which can be classified as Crohn’s disease (CD) and ulcerative colitis (UC) (1). IBD is a chronically relapsing and remitting condition of unknown origin that exhibits various features of immunological inflammation and affects at least 1 in 1,000 people in western countries. IBD is characterized by inflammation in the intestine, and is associated with diarrhea, occult blood, abdominal pain, weight loss, anemia and leukocytosis. IBD primarily affects young adults, and the disease initially manifests in childhood in 15–25% of cases. Therefore, IBD patients often develop severe symptoms that decrease their quality of life (2). Consequently, there is a need for innovative therapies for IBD.

Current treatments for IBD focus on suppressing inflammation or modulating the immune response using corticosteroids, mercaptopurines, 5-ASA, or monoclonal antibodies against inflammatory cytokines, e.g., the anti-tumor necrosis factor (TNF)-α antibody infliximab (3). However, despite the wide variety of pharmacologic options for patients with IBD, consistent cures and prolonged remissions have yet to be achieved.

Hepatocyte growth factor (HGF) was originally identified (47) and cloned (8,9) as a potent mitogen for hepatocytes, but has since been established as a multifunctional cytokine that exhibits mitogenic, motogenic, morphologic, angiogenic, antiapoptotic and organotrophic effects in a variety of tissues (10). HGF is upregulated in inflamed colonic mucosal tissue in patients with CD or UC (1113), and plasma HGF levels are elevated in animal models of acute colitis (14). In vitro, HGF modulates intestinal epithelial cell proliferation and migration (15), thereby enhancing epithelial cell restitution, which is the initial step of gastrointestinal wound healing. In addition, administration of recombinant human HGF (hHGF) protein reduces the severity of colitis and accelerates colonic mucosal repair in models of TNBS-induced and DSS-induced colitis (1619), as well as in HLA-B27 transgenic rats with colitis (20). Mukoyama et al (21) showed that the intrarectal administration of an adenoviral (Ad) vector carrying the HGF gene prevented TNBS-induced colitis. Additionally, Hanawa et al (22) demonstrated the attenuation of mouse DSS colitis by naked gene transfer of rat HGF into the liver, and Kanbe et al (23) reported the amelioration of mucosal damage in DSS colitis by the intrarectal administration of the naked HGF gene. In their study, Kanayama et al (24) demonstrated the promotion of colonic epithelial regeneration by HGF gene transfer through electroporation. Findings by those authors suggest that HGF is potentially an important new treatment modality for promoting the repair of intestinal mucosa in patients with IBD.

In the majority of previous studies, HGF was provided in the form of recombinant hHGF protein. However, due to the rapid clearance of the HGF protein, large doses and frequent administration of recombinant hHGF were required. Naked gene transfer is a simple and easy method, but the efficiency of gene transduction is extremely low, possibly leading to insufficient clinical effectiveness in human patients. By contrast, the intrarectal administration of an Ad carrying the HGF gene is considered to be extremely stressful for patients. Therefore, in this study we injected an Ad carrying the hHGF gene in single rounds of injections into both hindlimbs of mice 1 day after administration of DSS. We then investigated the therapeutic effects and mechanisms of systemically circulating HGF protein, produced by a gene introduced into the limbs, in the DSS-induced acute colitis model.

Materials and methods

Recombinant Ad

The Ad expressing hHGF under the transcriptional control of the cytomegalovirus immediate-early enhancer and a modified chicken β-actin promoter (Ad.HGF) was generated as described previously (25). The Ad.HGF and the control Ad expressing the LacZ gene (Ad.LacZ) were amplified in HEK-293 cells, purified twice on CsCl gradients, and desalted as described previously (2629).

Animal studies

Six- to 7-week-old female BALB/c mice weighing 17–20 g (Japan SLC, Inc., Hamamatsu, Japan) were housed in cages in a temperature-controlled environment under a 12-h light-dark cycle with free access to food and water. The animal studies were performed in accordance with the National Institutes of Health guidelines, as specified by the Animal Care Facility at Gifu University School of Medicine.

To induce dextran sodium sulfate (DSS) colitis, the mice were provided with distilled drinking water containing 5% (w/v) DSS (MW, 36,000–50,000; ICN Biomedicals Inc., Aurora, OH, USA) for 7 days. Subsequently, colitis was maintained by feeding the mice 1% DSS (3032) in the drinking water.

One day after the administration of DSS, Ad.HGF was injected into both hindlimbs of each mouse for a total dose of 1×1011 particles/mouse (i.e., 5×1010 particles each into the left and right thigh muscles) (n=8). Ad.LacZ was injected in a similar manner into control mice (n=8). These groups were followed until day 15 (i.e., 8 days after the end of the 7-day period of 5% DSS administration). To evaluate the severity of colitis, body weight was examined on a daily basis. On day 15, all the mice were sacrificed by inhaled anesthetics, and colon samples were collected for examination. In other experiments, on day 5 of 5% DSS administration, 5-bromo-2′-deoxyuridine (BrdU, 100 mg/kg) was administered intraperitoneally to mice (n=8) infected with Ad.HGF or Ad.LacZ, and the animals were sacrificed by inhaled anesthetics 2 h later. These samples were used for analyses of HGF signal transduction, cell proliferation, apoptosis, cytokines and lymphocyte surface markers. The concentration of exogenous hHGF in serum was analyzed using the same dose (i.e., 1×1011 particles/mouse) of Ad.LacZ or Ad.HGF in intact mice (n=16).

Enzyme-linked immunosorbent assay

The plasma concentration of hHGF following adenoviral intramuscular gene transduction (IMGT) was measured in mice at each time point (n=4) using the Quantikine human HGF Immunoassay kit (R&D Systems, Inc., Minneapolis, MN, USA). TNF-α, interleukin (IL)-1β, IL-6, interferon (IFN)-γ, IL-2, IL-4 and IL-5 levels in the colons of colitic mice were measured using commercially available enzyme-linked immunosorbent assay (ELISA) kits (BioSource International, Inc., Camarillo, CA, USA) according to the manufacturer’s instructions.

Immunoprecipitation and c-Met receptor phosphorylation assay

The phosphorylation and activation of the c-Met receptor in colon tissues were detected by immunoprecipitation, as described previously (33,34). In brief, 1 g of colon tissue was homogenized in 4 ml of lysis buffer [1% Triton X-100, 150 mM NaCl, 50 mM Tris-HCl (pH 7.6), 10% glycerol, 1 mM vanadate, and 1 mM phenylmethylsulfonyl fluoride] with a protease-inhibitor cocktail (Sigma-Aldrich, Tokyo, Japan). Following centrifugation, the supernatant was incubated with 0.5 μg/ml anti-mouse c-Met antibody (sc-162; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) for 4 h, and then sequentially incubated with 5 μl of protein G-Sepharose beads for 3 h. After washing, proteins bound to the beads were dissolved in sample buffer and subjected to SDS-PAGE. Phosphorylated c-Met was immunoblotted using the anti-phosphotyrosine antibody PY20 (Transduction Laboratories, Lexington, KY, USA).

Histopathological analysis

After each mouse was sacrificed, the intestine was dissected from the anus to the cecum and rinsed with physiological saline. The colon length was measured, and the colon sample was divided into three sections (cecum, proximal colon and distal colon), with the cecum being separated first, and then the remaining part of the colon being divided into two equal segments (proximal colon and distal colon). The cecum, proximal colon and distal colon were opened longitudinally, and the proximal and distal colon were equally divided longitudinally and transversely. Thus, the cecum was divided into two sections, and the proximal and distal colon were divided into four sections. The colon tissues were fixed in 10% formalin and embedded in paraffin, and 4-μm sections were cut and stained with hematoxylin and eosin (H&E) to determine the inflammation and crypt scores (35). Briefly, the sections were graded on a scale of 0–3 to indicate the severity of inflammation: 0, none; 1, mucosa; 2, mucosa and submucosa; and 3, transverse, and on a scale of 0–4 to indicate the severity of crypt damage: 0, none; 1, basal 1/3 damage; 2, basal 2/3 damage; 3: loss of the entire crypt with the surface epithelium remaining intact; and 4, loss of the entire crypt and surface epithelium. The changes were also scored with regard to the extent of tissue involvement, measured as a percentage: i) 1–25%, ii) 26–50%, iii) 51–75%, and iv) 76–100%. Each section was then separately scored for each feature by taking the product of the severity score and the score for the extent of tissue involvement. Thus, the inflammation score ranged from 0 to 12, and the crypt score ranged from 0 to 16. Apoptotic cells were detected using a light microscope (Olympus, Tokyo, Japan) and the terminal deoxynucleotidyltransferase-mediated deoxyuridine triphosphate biotin nick end-labeling (TUNEL) assay (ApopTag kit; Intergen Co., Purchase, NY, USA), as described previously (25,33,36). To detect proliferating cells, BrdU incorporation was measured using a staining kit (Zymed Laboratories, Inc., South San Francisco, CA, USA) according to the manufacturer’s instructions.

Endothelial cells, CD4+ T lymphocytes, and CD8+ T lymphocytes were detected in situ using an anti-vWF antibody (Dako Cytomation Co., Ltd., Kyoto, Japan), anti-CD4 antibody and anti-CD8 antibody (both from Zymed Laboratories, Inc.), respectively, as described previously (25,36).

Statistical analysis

Values provided are the means ± SEM values. The significance of differences was evaluated using the Student’s t-test.

Results

Intramuscular injection of Ad.HGF produces circulating plasma hHGF, leading to c-Met activation in the colonic mucosa

DSS-induced colitis was induced in 6- to 7-week-old female BALB/c mice. One day after DSS administration, Ad.HGF was administered in a single procedure involving injections into both hindlimbs (total dose, 1×1011 particles/mouse; as mentioned in Materials and methods). In the hHGF-overexpressing mice, the plasma levels of hHGF were 1,140±101, 634±341 and 33.9±15.8 pg/ml at 2, 4 and 6 days after injection, respectively. No hHGF was detected in the Ad.LacZ-treated mice at any time point, demonstrating that this method accurately detected only hHGF protein expressed from the hHGF transgene, without a cross-reaction resulting in detection of the endogenous mouse HGF protein. These results indicate that hHGF expression was effectively induced by the intramuscular injection of Ad.HGF, leading to the presence of hHGF in the plasma of the mice.

The biological effects of HGF are mediated by its receptor c-Met, which is capable of activating multiple intracellular transducers and signaling pathways. Therefore, we examined c-Met tyrosine phosphorylation in the colonic mucosal epithelium by western blotting (Fig. 1). Phosphorylated c-Met was detected at low or moderate levels in the injured colonic mucosa of mice treated with Ad.LacZ, presumably as a result of a DSS-induced increase in endogenous HGF in response to colonic mucosal injury (14). By contrast, the injured colonic mucosa of mice treated with Ad.HGF exhibited high levels of c-Met tyrosine phosphorylation.

Adenoviral hHGF IMGT prevents weight loss in DSS-induced colitis mice

DSS-induced colitis is characterized by bloody stools and severe weight loss (30). In mice treated with Ad.LacZ, we observed persistent liquid stool and waste with subsequent severe weight loss. By contrast, colitic mice that received a single round of injections of Ad.HGF exhibited significant reductions in liquid stool and gross bleeding from the rectum (data not shown). Fig. 2 shows the mean weight change, and that the body weights of Ad.HGF-treated mice were significantly higher than those of the Ad.LacZ-treated mice. In the Ad.LacZ-treated control mice, weight loss occurred 6–7 days after the initiation of DSS administration. Ad.HGF treatment significantly prevented this weight loss.

Adenoviral hHGF IMGT reduces colitis-induced intestinal shortening and pathological scores

Shortening of the colon correlates well with histologic changes, and colon length is therefore frequently used as a morphologic parameter to indicate the degree of inflammation (35). The colon lengths of mice treated with Ad.LacZ and Ad.HGF were 72.0±10.6 and 82.0±4.7 mm, respectively (Fig. 3A). In contrast to the colons in the Ad.HGF-treated group, the colons in the Ad.LacZ-treated group were short and severely inflamed, with evident hemorrhages (Fig. 3B).

To validate this finding, we evaluated the effect of Ad.HGF on DSS-induced colonic mucosal injury in mice by histological analysis at day 15. In the cecum and proximal part of the colon (i.e., towards the end of the cecum), the inflammation and crypt scores appeared to be decreased by Ad.HGF administration although this difference was not statistically significant (Figs. 4A and B, 5A and B). By contrast, treatment with Ad.HGF significantly decreased the inflammation and crypt scores in the distal part (i.e., towards the anus) and in the colon overall (Figs. 4C and D, 5C and D).

Kinetics of inflammation in colitic mice

To elucidate the mechanism underlying the therapeutic effect of hHGF, we studied the expression of TNF-α and IL-1β in the colon and evaluated the inflammation and crypt scores at days 4, 7, 10 and 14 of the experimental colitis model (Fig. 6). The expression of TNF-α and IL-1β peaked as early as day 4 (Fig. 6A and B). The inflammation and crypt scores peaked as early as day 7 (Fig. 6C and D). Given that the plasma concentration of hHGF protein peaked on day 2 and decreased thereafter, colon tissue were sampled and hHGF functions were analyzed on day 5.

Adenoviral hHGF IMGT suppresses apoptosis and enhances regeneration of the colonic epithelium

In DSS-induced colitis, loss of colonic mucosal epithelial cells is closely associated with apoptosis (37,38). To evaluate the role of Ad.HGF in preventing apoptosis in colonic epithelial cells, we performed the TUNEL assay to detect apoptotic cells (Fig. 7A). Ad.HGF-treated colitic mice had significantly (2.1-fold) fewer TUNEL-positive cells per high-power field (HPF) than Ad.LacZ-treated colitic mice.

To determine whether Ad.HGF-injection stimulated the proliferation of colonic epithelial cells, we measured the DNA labeling index in the colonic mucosal epithelium. As shown in Fig. 7B, the average number of BrdU-positive cells in the colonic mucosal epithelium was significantly (1.8-fold) higher in Ad.HGF-treated as compared to Ad.LacZ-treated mice, suggesting that hHGF stimulates proliferation in the colonic epithelial cells of colitic mice. These results suggested that adenoviral hHGF IMGT promoted survival and regeneration of the colonic mucosal epithelium in mice with DSS-induced colitis. HGF is known to promote angiogenesis (10). Therefore, we hypothesized that the angiogenic effect of HGF may contribute to the repair of the damaged colonic epithelium. However, when we analyzed angiogenesis in the distal part of the colon by anti-vWF immunohistochemistry, the number of blood vessels in the colon did not differ significantly between Ad.HGF-treated mice and controls, although a few more vessels appeared to be present in Ad.HGF-treated animals (Fig. 7C).

Effects of adenoviral hHGF IMGT on immunoreactive cells and inflammatory cytokines in DSS-induced colitis

To determine whether IMGT of hHGF affected the immune system of DSS-treated mice, we directly detected immune cells in the colon. Adenoviral hHGF IMGT decreased the number of CD4+ T cells and the CD4/CD8 ratio, but not the number of CD8+ T cells (Fig. 8).

The inflammatory cytokine cascade plays an important role in the pathogenesis of DSS-induced colitis. Therefore, we analyzed the cytokine profile of the entire colon by ELISA. In general, we observed upregulation of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in the colitic mice (39,40). The expression levels of TNF-α, IL-1β and IL-6 were further increased by hHGF IMGT (Fig. 9).

We also examined the effect of hHGF IMGT on Th1 (IFN-γ and IL-2) and Th2 (IL-4 and IL-5) cytokine expression in the colons of colitic mice. IFN-γ, IL-2 and IL-4 were upregulated by hHGF treatment (Fig. 10).

Discussion

This study evaluated the therapeutic potential of the intramuscular injection of HGF-expressing Ad for treating IBD, using a mouse model of DSS-induced colitis. The therapeutic strategy of adenoviral HGF IMGT, in which hHGF protein was produced at distal sites (hindlimbs) and systemically delivered to the target organ (the injured colon epithelium), functioned well. Epithelial cell injury in DSS-induced colitis was potently prevented by this method, which is clinically feasible, less invasive, and does not suffer from the drawbacks associated with the direct treatment of colitic tissues. Although previous studies (1618) have shown that HGF exerts protective effects in bowel disease, the regimens tested involved high levels of recombinant HGF protein (>100 μg/kg) and repeated injections.

Recent advances in molecular techniques have provided several strategies for in vivo gene delivery, including naked plasmid DNA, liposomes encapsulating DNA, and viral vectors (41,42). For instance, Hanawa et al (22) reported that administration of the naked HGF gene into the liver attenuated acute colitis in mice, and Kanbe et al (23) showed that intrarectal administration of a plasmid carrying the HGF gene ameliorated DSS-induced colitis in mice. Kanayama et al (24) found that colonic epithelial regeneration is promoted by HGF gene transfer via electroporation. Oh et al (43) reported that HVJ liposomes encapsulating the hHGF gene ameliorated TNBS-induced colitis in mice, and that intrarectal administration of an Ad carrying the HGF gene improved colonic damage in TNBS-induced colitis (21). However, each type of gene therapy system used thus far has some associated limitations and concerns, particularly from the viewpoints of clinical applicability, feasibility and safety (41,42).

In this study, we assessed for the first time the therapeutic potential of a unique method of adenoviral hHGF IMGT for treating IBDs. In accordance with the results obtained in our previous studies of a mouse model of myocardial infarction (25,36), we successfully detected circulating hHGF in the plasma of colitic mice after adenoviral hHGF IMGT. In the colons of colitic mice that received adenoviral hHGF IMGT, the c-Met/HGF receptor was highly phosphorylated on tyrosine, demonstrating the functional efficacy of the adenoviral hHGF IMGT system. Furthermore, hHGF IMGT stimulated proliferation and inhibited apoptosis in the disrupted intestinal epithelial barrier. These results indicate that our hHGF IMGT system induces protection and regeneration in the colon, suggesting that it would be useful in clinical treatments for bowel diseases.

The effects of HGF on carcinogenesis remain unclear. Some studies suggest that HGF may promote the growth and metastasis of some cancer types, probably via the stimulation of cancer cell growth and angiogenesis (44,45). By contrast, carcinogenesis or malignant phenotypes in other cancer types are potently inhibited by overexpressed HGF (33). The effects of HGF on IBDs are also unclear. In general, tumor development may be caused by long-term exposure of cells to an abnormally overexpressed growth factor. In our therapeutic system, the duration of hHGF secretion after single rounds of intramuscular injection was relatively short; therefore, we consider the risk of cancer occurrence to be very low. In addition, a previous study demonstrated the efficacy of repeated administration of Ad into muscles, suggesting that this approach may yield sustained and elevated therapeutic efficiency: neutralizing antibodies against adenovirus should hinder only Ad circulating in the bloodstream, but not Ad administered into the muscle (46). These findings are encouraging with regard to the potential safety and clinical applicability of this approach.

With regard to the therapeutic mechanism, previous studies have reported that administration of recombinant HGF protein (16) and vector encoding HGF gene (43) ameliorate TNBS-induced colitis and reduced inflammation, decreasing the levels of inflammatory cytokines such as TNF-α. In particular, Oh et al (43) showed that administration of a plasmid carrying the HGF gene reduced the invasion of CD4+ cells and neutrophils and suppressed the expression of Th1 cytokines such as IL-12, IL-1β and IFN-γ in a TNBS-induced colitis model. Hanawa et al (22) showed that administration of an HGF gene-containing plasmid in the liver by intravenous injection suppressed the mRNA levels of IFN-γ, IL-18 and TNF-α, and increased the mRNA levels of anti-inflammatory cytokines such as IL-10. Jeschke et al (47) found that recombinant HGF reduced burn-related damage to the small intestine. The serum levels of TNF-α, IL-1β and IL-6 were higher in the HGF-treated group than in the control group. However, Jeschke et al (47) did not explain why the levels of these cytokines were increased by HGF. Our data indicate that the number of CD4+ cells decreased, but the levels of TNF-α, IL-1β and IL-6, as well as those of Th1 and Th2 cytokines such as IL-2, IFN-γ and IL-4, were elevated in the Ad.HGF-treated group. We hypothesize that the reasons for the differences between our findings and those of previous studies may involve differences among mouse strains, our use of intramuscular gene administration mediated by an Ad, and our selection of the early phase of DSS colitis for analysis of inflammation and cytokine expression.

Futamatsu et al (48) reported that HGF suppressed T-cell proliferation and IFN-γ production and increased IL-4 and IL-10 secretion from CD4+ T cells in vitro, and also reduced the severity of experimental autoimmune myocarditis in vivo by inducing Th2 cytokines and suppressing apoptosis of cardiomyocytes. Kuroiwa et al (49) demonstrated that HGF gene delivery inhibited Th2 immune responses and ameliorated lupus nephritis, autoimmune sialadenitis, and cholangitis in chronic GVHD mice. Another study indicated that treatment with HGF potently suppressed dendritic cell functions such as antigen-presenting capacity, both in vitro and in vivo, thus downregulating antigen-induced Th1 and Th2 immune responses in a mouse model of allergic airway inflammation (50). HGF has been suggested to suppress airway hyper-responsiveness, inflammation, remodeling, and eosinophil function in asthma (51). Okunishi et al (52) reported that HGF suppresses antigen-induced T-cell priming by regulating the functions of dendritic cells through IL-10 downregulation in the antigen-sensitization phase. By contrast, they found that repeated treatment with HGF induced Th2 immune responses with the upregulation of IL-10 by DCs in the chronic inflammation phase of a mouse model of collagen-induced arthritis. Thus, it is clear that HGF induces various immune responses in different disease models. However, further analysis is required to clarify the effects of HGF on the immune system.

In conclusion, we have shown that a single round of intramuscular injections of adenoviral hHGF is sufficient to inhibit apoptosis and reconstitute the epithelium in a mouse model of DSS-induced colitis. Based on these results, this approach shows promise for clinical application in IBD.

Acknowledgements

This study was supported in part by a Grant-in-Aid for Scientific Research (C) from the Ministry of Education, Culture, Sports, Science and Technology of Japan. We would like to thank Hatsue Oshika for her technical assistance.

References

1 

Hisamatsu T, Kanai T, Mikami Y, Yoneno K, Matsuoka K and Hibi T: Immune aspects of the pathogenesis of inflammatory bowel disease. Pharmacol Ther. 137:283–297. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bernklev T, Jahnsen J, Aadland E, et al: Health-related quality of life in patients with inflammatory bowel disease five years after the initial diagnosis. Scand J Gastroenterol. 39:365–373. 2004.PubMed/NCBI

3 

Burger D and Travis S: Conventional medical management of inflammatory bowel disease. Gastroenterology. 140:1827–1837. e18222011. View Article : Google Scholar : PubMed/NCBI

4 

Nakamura T, Nawa K and Ichihara A: Partial purification and characterization of hepatocyte growth factor from serum of hepatectomized rats. Biochem Biophys Res Commun. 122:1450–1459. 1984. View Article : Google Scholar : PubMed/NCBI

5 

Russell WE, McGowan JA and Bucher NL: Partial characterization of a hepatocyte growth factor from rat platelets. J Cell Physiol. 119:183–192. 1984. View Article : Google Scholar : PubMed/NCBI

6 

Thaler FJ and Michalopoulos GK: Hepatopoietin A: partial characterization and trypsin activation of a hepatocyte growth factor. Cancer Res. 45:2545–2549. 1985.PubMed/NCBI

7 

Gohda E, Tsubouchi H, Nakayama H, et al: Human hepatocyte growth factor in plasma from patients with fulminant hepatic failure. Exp Cell Res. 166:139–150. 1986. View Article : Google Scholar : PubMed/NCBI

8 

Nakamura T, Nishizawa T, Hagiya M, et al: Molecular cloning and expression of human hepatocyte growth factor. Nature. 342:440–443. 1989. View Article : Google Scholar : PubMed/NCBI

9 

Miyazawa K, Tsubouchi H, Naka D, et al: Molecular cloning and sequence analysis of cDNA for human hepatocyte growth factor. Biochem Biophys Res Commun. 163:967–973. 1989. View Article : Google Scholar : PubMed/NCBI

10 

Matsumoto K and Nakamura T: Hepatocyte growth factor (HGF) as a tissue organizer for organogenesis and regeneration. Biochem Biophys Res Commun. 239:639–644. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Matsuno M, Shiota G, Umeki K, Kawasaki H, Kojo H and Miura K: Clinical evaluation of hepatocyte growth factor in patients with gastrointestinal and pancreatic diseases with special reference to inflammatory bowel disease. Res Commun Mol Pathol Pharmacol. 97:25–37. 1997.PubMed/NCBI

12 

Kitamura S, Kondo S, Shinomura Y, et al: Expression of hepatocyte growth factor and c-met in ulcerative colitis. Inflamm Res. 49:320–324. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Srivastava M, Zurakowski D, Cheifetz P, Leichtner A and Bousvaros A: Elevated serum hepatocyte growth factor in children and young adults with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 33:548–553. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Ortega-Cava CF, Ishihara S, Kawashima K, et al: Hepatocyte growth factor expression in dextran sodium sulfate-induced colitis in rats. Dig Dis Sci. 47:2275–2285. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Dignass AU, Lynch-Devaney K and Podolsky DK: Hepatocyte growth factor/scatter factor modulates intestinal epithelial cell proliferation and migration. Biochem Biophys Res Commun. 202:701–709. 1994. View Article : Google Scholar : PubMed/NCBI

16 

Numata M, Ido A, Moriuchi A, et al: Hepatocyte growth factor facilitates the repair of large colonic ulcers in 2,4,6-trinitrobenzene sulfonic acid-induced colitis in rats. Inflamm Bowel Dis. 11:551–558. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Tahara Y, Ido A, Yamamoto S, et al: Hepatocyte growth factor facilitates colonic mucosal repair in experimental ulcerative colitis in rats. J Pharmacol Exp Ther. 307:146–151. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Ohda Y, Hori K, Tomita T, et al: Effects of hepatocyte growth factor on rat inflammatory bowel disease models. Dig Dis Sci. 50:914–921. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Setoyama H, Ido A, Numata M, et al: Repeated enemas with hepatocyte growth factor selectively stimulate epithelial cell proliferation of injured mucosa in rats with experimental colitis. Life Sci. 89:269–275. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Arthur LG, Schwartz MZ, Kuenzler KA and Birbe R: Hepatocyte growth factor treatment ameliorates diarrhea and bowel inflammation in a rat model of inflammatory bowel disease. J Pediatr Surg. 39:139–143. 2004. View Article : Google Scholar

21 

Mukoyama T, Kanbe T, Murai R, et al: Therapeutic effect of adenoviral-mediated hepatocyte growth factor gene administration on TNBS-induced colitis in mice. Biochem Biophys Res Commun. 329:1217–1224. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Hanawa T, Suzuki K, Kawauchi Y, et al: Attenuation of mouse acute colitis by naked hepatocyte growth factor gene transfer into the liver. J Gene Med. 8:623–635. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Kanbe T, Murai R, Mukoyama T, et al: Naked gene therapy of hepatocyte growth factor for dextran sulfate sodium-induced colitis in mice. Biochem Biophys Res Commun. 345:1517–1525. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Kanayama M, Takahara T, Yata Y, et al: Hepatocyte growth factor promotes colonic epithelial regeneration via Akt signaling. Am J Physiol Gastrointest Liver Physiol. 293:G230–G239. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Li Y, Takemura G, Kosai K, et al: Postinfarction treatment with an adenoviral vector expressing hepatocyte growth factor relieves chronic left ventricular remodeling and dysfunction in mice. Circulation. 107:2499–2506. 2003. View Article : Google Scholar

26 

Chen SH, Chen XH, Wang Y, et al: Combination gene therapy for liver metastasis of colon carcinoma in vivo. Proc Natl Acad Sci USA. 92:2577–2581. 1995. View Article : Google Scholar : PubMed/NCBI

27 

Takahashi T, Kawai T, Ushikoshi H, et al: Identification and isolation of embryonic stem cell-derived target cells by adenoviral conditional targeting. Mol Ther. 14:673–683. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Okabe Y, Kusaga A, Takahashi T, et al: Neural development of methyl-CpG-binding protein 2 null embryonic stem cells: a system for studying Rett syndrome. Brain Res. 1360:17–27. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Horikawa Y, Wang Y, Nagano S, et al: Assessment of an altered E1B promoter on the specificity and potency of triple-regulated conditionally replicating adenoviruses: implications for the generation of ideal m-CRAs. Cancer Gene Ther. 18:724–733. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y and Nakaya R: A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 98:694–702. 1990.PubMed/NCBI

31 

Tomoyose M, Mitsuyama K, Ishida H, Toyonaga A and Tanikawa K: Role of interleukin-10 in a murine model of dextran sulfate sodium-induced colitis. Scand J Gastroenterol. 33:435–440. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Kanauchi O, Nakamura T, Agata K, Mitsuyama K and Iwanaga T: Effects of germinated barley foodstuff on dextran sulfate sodium-induced colitis in rats. J Gastroenterol. 33:179–188. 1998. View Article : Google Scholar : PubMed/NCBI

33 

Yuge K, Takahashi T, Nagano S, et al: Adenoviral gene transduction of hepatocyte growth factor elicits inhibitory effects for hepatoma. Int J Oncol. 27:77–85. 2005.PubMed/NCBI

34 

Kamisasanuki T, Tokushige S, Terasaki H, et al: Targeting CD9 produces stimulus-independent antiangiogenic effects predominantly in activated endothelial cells during angiogenesis: a novel antiangiogenic therapy. Biochem Biophys Res Commun. 413:128–135. 2011. View Article : Google Scholar

35 

Murthy SN, Cooper HS, Shim H, Shah RS, Ibrahim SA and Sedergran DJ: Treatment of dextran sulfate sodium-induced murine colitis by intracolonic cyclosporin. Dig Dis Sci. 38:1722–1734. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Li Y, Takemura G, Kosai K, et al: Critical roles for the Fas/Fas ligand system in postinfarction ventricular remodeling and heart failure. Circ Res. 95:627–636. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Iwamoto M, Koji T, Makiyama K, Kobayashi N and Nakane PK: Apoptosis of crypt epithelial cells in ulcerative colitis. J Pathol. 180:152–159. 1996. View Article : Google Scholar : PubMed/NCBI

38 

Sträter J, Wellisch I, Riedl S, et al: CD95 (APO-1/Fas)-mediated apoptosis in colon epithelial cells: a possible role in ulcerative colitis. Gastroenterology. 113:160–167. 1997.PubMed/NCBI

39 

Rogler G and Andus T: Cytokines in inflammatory bowel disease. World J Surg. 22:382–389. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Egger B, Bajaj-Elliott M, MacDonald TT, Inglin R, Eysselein VE and Büchler MW: Characterisation of acute murine dextran sodium sulphate colitis: cytokine profile and dose dependency. Digestion. 62:240–248. 2000. View Article : Google Scholar : PubMed/NCBI

41 

Tomanin R and Scarpa M: Why do we need new gene therapy viral vectors? Characteristics, limitations and future perspectives of viral vector transduction. Curr Gene Ther. 4:357–372. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Boulaiz H, Marchal JA, Prados J, Melguizo C and Aránega A: Non-viral and viral vectors for gene therapy. Cell Mol Biol (Noisy-le-grand). 51:3–22. 2005.

43 

Oh K, Iimuro Y, Takeuchi M, et al: Ameliorating effect of hepatocyte growth factor on inflammatory bowel disease in a murine model. Am J Physiol Gastrointest Liver Physiol. 288:G729–G735. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Shiota G, Kawasaki H, Nakamura T and Schmidt EV: Characterization of double transgenic mice expressing hepatocye growth factor and transforming growth factor alpha. Res Commun Mol Pathol Pharmacol. 90:17–24. 1995.PubMed/NCBI

45 

Sakata H, Takayama H, Sharp R, Rubin JS, Merlino G and LaRochelle WJ: Hepatocyte growth factor/scatter factor overexpression induces growth, abnormal development, and tumor formation in transgenic mouse livers. Cell Growth Differ. 7:1513–1523. 1996.

46 

Chen P, Kovesdi I and Bruder JT: Effective repeat administration with adenovirus vectors to the muscle. Gene Ther. 7:587–595. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Jeschke MG, Bolder U, Finnerty CC, et al: The effect of hepatocyte growth factor on gut mucosal apoptosis and proliferation, and cellular mediators after severe trauma. Surgery. 138:482–489. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Futamatsu H, Suzuki J, Mizuno S, et al: Hepatocyte growth factor ameliorates the progression of experimental autoimmune myocarditis: a potential role for induction of T helper 2 cytokines. Circ Res. 96:823–830. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Kuroiwa T, Iwasaki T, Imado T, Sekiguchi M, Fujimoto J and Sano H: Hepatocyte growth factor prevents lupus nephritis in a murine lupus model of chronic graft-versus-host disease. Arthritis Res Ther. 8:R1232006. View Article : Google Scholar : PubMed/NCBI

50 

Okunishi K, Dohi M, Nakagome K, et al: A novel role of hepatocyte growth factor as an immune regulator through suppressing dendritic cell function. J Immunol. 175:4745–4753. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Ito W, Takeda M, Tanabe M, et al: Anti-allergic inflammatory effects of hepatocyte growth factor. Int Arch Allergy Immunol. 146(Suppl 1): S82–S87. 2008. View Article : Google Scholar

52 

Okunishi K, Dohi M, Fujio K, et al: Hepatocyte growth factor significantly suppresses collagen-induced arthritis in mice. J Immunol. 179:5504–5513. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2014
Volume 33 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yuge K, Takahashi T, Khai NC, Goto K, Fujiwara T, Fujiwara H and Kosai K: Intramuscular injection of adenoviral hepatocyte growth factor at a distal site ameliorates dextran sodium sulfate-induced colitis in mice. Int J Mol Med 33: 1064-1074, 2014
APA
Yuge, K., Takahashi, T., Khai, N.C., Goto, K., Fujiwara, T., Fujiwara, H., & Kosai, K. (2014). Intramuscular injection of adenoviral hepatocyte growth factor at a distal site ameliorates dextran sodium sulfate-induced colitis in mice. International Journal of Molecular Medicine, 33, 1064-1074. https://doi.org/10.3892/ijmm.2014.1686
MLA
Yuge, K., Takahashi, T., Khai, N. C., Goto, K., Fujiwara, T., Fujiwara, H., Kosai, K."Intramuscular injection of adenoviral hepatocyte growth factor at a distal site ameliorates dextran sodium sulfate-induced colitis in mice". International Journal of Molecular Medicine 33.5 (2014): 1064-1074.
Chicago
Yuge, K., Takahashi, T., Khai, N. C., Goto, K., Fujiwara, T., Fujiwara, H., Kosai, K."Intramuscular injection of adenoviral hepatocyte growth factor at a distal site ameliorates dextran sodium sulfate-induced colitis in mice". International Journal of Molecular Medicine 33, no. 5 (2014): 1064-1074. https://doi.org/10.3892/ijmm.2014.1686