Open Access

Matrix metalloproteinase 14 participates in corneal lymphangiogenesis through the VEGF-C/VEGFR-3 signaling pathway

  • Authors:
    • Hai‑Tao Du
    • Ping Liu
  • View Affiliations

  • Published online on: August 22, 2016     https://doi.org/10.3892/etm.2016.3601
  • Pages: 2120-2128
  • Copyright: © Du et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The aim of the present study was to investigate the roles of matrix metalloproteinase 14 (MMP-14) in corneal inflammatory lymphangiogenesis. The expression of MMP-14 in vivo was detected by immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot assays, under various corneal conditions. pCMV‑MMP‑14 or empty pCMV vectors were injected into mouse corneal stroma, 3 days after suture placement in a standard suture‑induced inflammatory corneal neovascularization assay. The outgrowth of blood and lymphatic vessels and macrophage recruitment were analyzed using immunofluorescence. The expression levels of vascular endothelial growth factor (VEGF) subtypes were tested by RT-qPCR. MMP‑14 expression was upregulated significantly following various corneal injuries. The results demonstrated, for the first time, that MMP‑14 strongly promotes corneal lymphangiogenesis and macrophage infiltration during inflammation. Furthermore, expression levels of VEGF‑C and VEGF receptor‑3, but not other VEGF components, were significantly upregulated by the intrastromal delivery of MMP‑14 during corneal lymphangiogenesis. In conclusion, this study indicates that MMP‑14 is critically involved in the processes of lymphangiogenesis. Inhibition of MMP‑14 may provide a viable treatment for transplant rejection and other lymphatic disorders.

Introduction

The healthy cornea is devoid of blood and lymphatic vessels. However, vascularization of the cornea can occur during a number of pathological disorders, such as corneal chronic inflammation, alkali burns and graft rejection (1,2). The outgrowth of blood and lymphatic vessels from the limbus into the cornea, which is defined as corneal hemangiogenesis (HG) and lymphangiogenesis (LG), reduces transparency and visual acuity (3,4). It is also widely recognized that corneal HG and LG are strong risk factors for graft failure. Studies have shown that inhibition of corneal neovascularization (NV) can promote graft survival in a murine model of corneal transplantation (5,6). Thus, inhibition of corneal NV is a key point for not only optimal clarity and vision, but also higher graft survival rates.

Unlike blood vessels, corneal lymphatic vessels are not easily visible; therefore, systematic lymphatic research started much later than the study of blood vessels. With the advancement of technologies and the discovery of several lymphatic endothelial-specific markers, such as lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), prospero homeobox protein 1 (Prox-1) and vascular endothelial growth factor receptor-3 (VEGFR-3), significant progress has been made (79). A number of factors involved in LG have been identified to date, including VEGF-C and VEGF-D (1012); however, their underlying mechanisms remain unknown.

Matrix metalloproteinases (MMPs), which are a family of zinc-dependent enzymes, play key roles in degrading the extracellular matrix (ECM) associated with metastasis, angiogenesis and tumor invasion (13,14). MMP-14 (also known as membrane type-1 matrix metalloproteinase), a transmembrane type MMP, has been reported to possess a broad spectrum of activity against ECM components such as type I and II collagen, fibronectin, vitronectin, laminin, fibrin and proteoglycans (15,16). A previous study suggested that MMP-14 also enhanced cancer cell migration and invasion by the shedding of cluster of differentiation (CD)44 and syndecan-1 from the cell surface, which indicated that MMP-14 participated in tumor invasion and metastasis (17). MMP-14 may promote angiogenesis by degrading the ECM (18), and may also regulate corneal HG by cleaving decorin, an anti-angiogenic factor in corneas with basic fibroblast growth factor (bFGF)-induced vascularization (19). Furthermore, MMP-14 expression has been shown to be enhanced in wounded corneal epithelium and stroma (20). In contrast to HG, LG is known to also participate in corneal injuries, but the contribution of MMP-14 to this process remains unclear.

Hence, the present study aimed to determine if MMP-14 promotes corneal LG, in vivo. The expression of MMP-14, which is able to induce the outgrowth of lymphatic vessels after various corneal injuries, was determined by immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot assays. In addition, whether corneal LG was increased in a murine model of suture-induced inflammatory corneal NV was investigated, by the intrastromal injection of MMP-14 plasmid. Finally, whether VEGF signaling was involved in the molecular pathway leading to corneal LG was investigated, through the intrastromal delivery of MMP-14.

Materials and methods

Plasmid construction

A cDNA encoding the MMP-14 gene was subcloned into the pCMV vector (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) as previously described (21). Plasmids were purified using a Qiagen plasmid purification kit (Qiagen, Inc., Santa Clarita, CA, USA) according to the manufacturer's protocol. The concentration of plasmid was ~2 µg/µl for naked DNA injection.

Animals and anesthesia

All animal protocols were approved by the local animal care committee of the First Affiliated Hospital of Harbin Medical University (Harbin, China), and were in accordance with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. A total of 153 male C57BL/6 mice (6–8 weeks old; 18–20 g) were purchased from the Laboratory Animal Center of Harbin Medical University (Harbin, China). All the mice were raised in constant room temperature and free for food and water. The room was in 12-h light/dark cycle. Mice were anesthetized with an intraperitoneal injection of a combination of ketamine and xylazine (120 and 20 mg/kg bodyweight, respectively). The mice were sacrificed by CO2 inhalation overdose at the end of the experiment.

Suture-induced corneal NV and corneal intrastromal injections

The mouse model of suture-induced inflammatory corneal NV was established as previously described (22). The central cornea was marked with a 2-mm diameter trephine and three 11–0 nylon sutures (Lingqiao; Ningbo Medical Needle Co., Ltd., Ningbo, China) were placed in the intrastromal position. The outer point of suture placement was near the limbus, and the inner point was near the corneal center equidistant from the limbus. Sutures were removed after 7 days. Corneal intrastromal injections were performed on day 3 after suture placement, as previously described (23). A 0.5-inch 33-gauge needle on a 10-µl gas tight syringe (Hamilton Robotics, Reno, NV, USA) was introduced into the corneal stroma, and plasmid (5 µg pCMV-MMP14 or pCMV) solution was injected into the stroma to separate corneal lamellae and disperse the plasmid. Thus, MMP-14 and empty vector (pCMV) groups were established. Normal mice were entirely untreated, and saline control mice received only standard suture placement, and treated eyes were then rinsed with sterile physiological saline (0.9% NaCl, 1 ml, twice daily for 1 week). A total of 35 male C57BL/6 mice were used, including 8 mice in untreated group, 7 mice in saline control group, 8 mice in empty vector group and 10 mice in the MMP-14 group.

Mouse alkali injury model

The alkali burn injury model in the mouse cornea was used, as described previously (n=9) (24). In brief, a 2-mm diameter filter paper disc was wetted with 1 mol/l NaOH solution for 20 sec and placed on the central cornea of the mouse for 30 sec. Injured eyes were rinsed with sterile physiological saline (0.9% NaCl, 20 ml) immediately.

Immunohistochemistry

Corneas were cut and fixed in 10% neutral buffered formalin for 24 h. Paraffin-embedded tissue sections (4 µm) were deparaffinized, rehydrated, and treated with 0.3% hydrogen peroxide in methanol for 30 min, to eliminate endogenous peroxidase activity. The tissue sections were then incubated for 60 min at room temperature with a rabbit anti-mouse MMP-14 monoclonal antibody (1:2,000; ab51074; Abcam, Cambridge, UK). After three washes (3 min each) with phosphate-buffered saline (PBS), a DAB Detection kit (PV9000; ZSGB-Bio, Beijing, China) was used for MMP-14 staining, according to the manufacturer's protocol. Images were acquired with the Leica DM4000B biological microscope equipped with a Leica DFC 550 digital camera and Leica Application Suite version 4.2.0 software (Leica Biosystems, GmbH, Heidelberg, Germany).

RNA isolation and RT-qPCR

Total RNA was extracted from the corneas using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Total RNA (400 ng) was reverse transcribed using the PrimeScript™ RT reagent kit with gDNA Eraser (Takara Biotechnology Co., Ltd., Dalian, China). qPCR was performed using SYBR® Premix Ex Taq™ II (Takara Biotechnology Co., Ltd.) with a LightCycler 480 Real-time PCR System (Roche Diagnostics, Basel, Switzerland). qPCR was performed under the following conditions: Initial denaturation step of 95°C for 30 sec, 40 cycles of 95°C for 5 sec, and of 60°C for 30 sec, followed by an additional denaturation step of 95°C for 5 sec and 60°C for 60 sec, as a subsequent melt curve analysis to check amplification specificity. All assays were conducted three times, and were performed in triplicate. Results were derived from the comparative threshold cycle method (25,26) and normalized by glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as an internal control. The primers used for qPCR were as shown in Table I.

Table I.

Primers used for quantitative polymerase chain reaction.

Table I.

Primers used for quantitative polymerase chain reaction.

GeneSequence (5′-3′)
GAPDHF: GTATTGGGCGCCTGGTCACC
R: CGCTCCTGGAAGATGGTGATGG
VEGF-AF: ACACGGTGGTGGAAGAAGAG
R: CAAGTCTCCTGGGGACAGAA
VEGF-CF: CTACAGATGTGGGGGTTGCT
R: GATTGGCAAAACTGATTGTGAC
VEGF-DF: GAGGCTGCTGCAACGAAGA
R: GCACTTACAACCCGTATGGTT
VEGFR-1F: CTGGACTGAGACCAAGCCCAAG
R: GCTCAGATTCATCGTCCTGCAC
VEGFR-2F: CTGTATGGAGGAAGAGGAAGTG
R: GGTTCCTCCAATGGGATATC
VEGFR-3F: CTCTGACCTAGTGGAGATCCTG
R: CTTCGGTGATATGTAGAGCTGTG
MMP-14F: GCTTTACTGCCAGCGTTC
R: CCCACTTATGGATGAAGCAAT

[i] F, forward; R, reverse; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MMP, matrix metalloproteinase.

Western blot analysis

The corneas were harvested and lysed in ice-cold radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Shanghai, China) with the addition of protease inhibitors. After separation by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, proteins were transferred onto nitrocellulose membranes. The membranes were incubated in blocking solution [2% bovine serum albumin (BSA) in Tris-buffered saline with Tween-20; Beyotime Institute of Biotechnology] for 1 h at room temperature, then incubated with a rabbit anti-mouse MMP-14 monoclonal antibody (1:2,000; ab51074; Abcam) overnight at 4°C. Each step was followed by extensive washing with PBS. The membranes were then incubated with corresponding horseradish peroxidase-conjugated secondary antibody (1:5,000; A0545; Sigma-Aldrich) for 1 h at room temperature, and developed using an enhanced chemiluminescence system (RPN2108; GE Healthcare, Little Chalfont, UK.). β-actin was used as loading control. The antibody was mouse anti-mouse β-actin monoclonal antibody (1:1,000; A00702-100; GenScript, Nanjing, China).

Corneal immunofluorescence microscopy and quantification

The immunofluorescence experiments were performed as previously described (27). The excised corneas were rinsed in PBS and fixed in acetone for 30 min. After washing and blocking with 2% BSA in PBS for 2 h, the corneas were stained overnight at 4°C with a rabbit anti-mouse LYVE-1 antibody (1:500; Abcam) and a rat anti-mouse CD31 antibody (1:100; BD Pharmingen, San Diego, CA, USA). On day 2, the tissue was washed three times with PBS, and was stored at 4°C in the absence of light. The LYVE-1 antibody (ab14917) was detected with an Alexa Fluor 647-conjugated goat anti-rabbit IgG antibody (1:200; A-21244; Invitrogen; Thermo Fisher Scientific, Inc.) and the CD31 antibody (550274) was detected with an Alexa Fluor 488-conjugated goat anti-rat IgG antibody (1:200; A-11006; Invitrogen; Thermo Fisher Scientific, Inc.). The corneas were stained with the Alexa Fluor antibodies overnight at 4°C. To detect the recruitment of macrophages into the inflamed corneas, a fluorescein isothiocyanate-conjugated rat anti-mouse CD11b antibody (1:100; 557396; BD Pharmingen) was used. The excised corneas were rinsed by PBS and fixed in acetone for 30 min. After washing and blocking with 2% BSA in PBS, for 2 h, corneas were stained overnight at 4°C, with a CD11b antibody.

The stained whole mounts were analyzed with a fluorescence microscope (EVOS f1; AMG; Thermo Fisher Scientific, Inc.). Each whole mount image was quantified using ImageJ software (National Institutes of Health, Bethesda, MD, USA), and the version was 1.24o. A detailed explanation of this method has been described previously (28). The mean vascularized area of the control groups was defined as being 100%; vascularized areas were then related to this value (vessel ratio). For macrophage analysis, the mean number of macrophages of the control groups was set as 100%; the numbers of macrophages per whole mount were then related to this value (cell ratio).

Statistical analysis

Statistical analysis was performed using Student's t-test with SPSS version 13.0 software (SPSS, Inc., Chicago, IL, USA). Results were expressed as the mean ± standard error of the mean, and a value of P<0.05 was considered to indicate a statistically significant difference. Graphs were drawn using GraphPad Prism, version 5.0 software (GraphPad Software, Inc., La Jolla, CA, USA).

Results

MMP-14 expression in the cornea

Immunohistochemical staining revealed weak expression of MMP-14 in normal cornea, and high expression levels of MMP-14 in the cornea on day 3 after standard suture placement or after alkali injury (Fig. 1A-C). As shown, these corneal injuries resulted in evident corneal NV. Moreover, the levels of corneal MMP-14 expression were also increased on the first day after the intrastromal injection of MMP-14 plasmid when compared with that of normal cornea, but increased levels were not detected in the vector-injected cornea. The data indicated that the intrastromal delivery of MMP-14 plasmid resulted in increased MMP-14 expression. The levels of MMP-14 expression were almost returned to normal at 3 days after the injection of MMP-14 DNA (Fig. 1D-F).

The results of the RT-qPCR assays demonstrated that MMP-14 expression was significantly upregulated in corneas treated with an intrastromal injection of MMP-14 plasmid (P=0.007), suture placement (P=0.003) or alkali injury (P=0.002), whereas MMP-14 expression in corneas treated with empty vector injection (P=0.442) or in corneas 3 days after MMP-14 vector injection (P=0.239) showed no significant changes, in comparison with the normal cornea (Fig. 2A). Furthermore, western blot assay confirmed the significant upregulation of MMP-14 expression in the aforementioned corneas (Fig. 2B).

Intrastromal delivery of MMP-14 DNA promotes corneal LG and HG following suture placement

The standard suture-induced corneal NV assay and corneal intrastromal injection model were used to investigate the effect of MMP-14. Mice were randomized to receive intrastromal injections of either pCMV-MMP-14 or pCMV empty vector on day 3 after suture placement. On day 7, the densities of CD31-positive blood vessels and LYVE-1-positive lymphatic vessels were detected by immunohistochemistry as described previously (29). Quantitative immunohistochemical and morphometric analyses clearly revealed that corneal LG and HG were induced by standard suture placement, and injection of MMP-14 led to a significant promotion of LG and HG (Fig. 3A-I). The numbers of lymphatic vessels in mice treated with MMP-14 were significantly increased (P=0.009), and the numbers of blood vessels were also markedly increased (P=0.011), in comparison with those in the mice treated with empty vector (Fig. 3J and K).

Treatment with MMP-14 promotes the recruitment of inflammatory macrophages into the cornea

To test whether the lymphangiogenic effect of MMP-14 was also partially caused by an indirect effect on macrophages, CD11b+ macrophage infiltration into inflamed corneas (n=7 mice) was evaluated using standard corneal suture placement and intrastromal injection models. Treatment with MMP-14 resulted in a significantly increased recruitment of CD11b+ compared with empty vector (P=0.019), as shown in Fig. 4.

Treatment with MMP-14 upregulates the expression of VEGF-C and VEGFR-3

HG and LG are driven by the production of lymphangiogenic growth factors, and the VEGF pathway is central to these processes (30). To further investigate the roles of MMP-14 in HG and LG, RT-qPCR was used to examine the expression of VEGF ligands and receptors. The results showed that VEGF-C (P=0.028) and VEGFR-3 (P=0.011) expression levels were significantly upregulated in mice treated with MMP-14, whereas the expression levels of VEGF-A (P=0.198), VEGF-D (P=0.183), VEGFR-1 (P=0.296) and VEGFR-2 (P=0.321) showed no significant changes, in comparison with those in the empty vector group (Fig. 5).

Discussion

For the first time, to the best of our knowledge, the present study provides evidence that MMP-14 promotes in vivo LG in a corneal suture-induced mouse model. Although previous studies have reported the effects of MMP-14 on several angiogenesis-related properties, including degradation of ECM and cleavage of decorin (18), its contribution during LG has received less attention and so further investigation is merited.

It has previously been shown that proMMP-2 activation can be blocked by a specific monoclonal antibody against MMP-14, which resulted in a marked reduction of lymphatic vessel sprouting (31). However, the impact of MMP-14 on LG in vivo was not determined in that study. In the present study, corneal LG and HG were significantly increased in the suture-induced inflammatory corneal NV model when naked MMP-14 DNA was added. Thus, it may be concluded that MMP-14 plays an important role in the development of new lymphatic vessels.

To assess the association between MMP-14 and corneal NV, MMP-14 expression was investigated under various corneal conditions using immunohistochemical analysis, RT-qPCR and western blot analysis. In the present study, corneal intrastromal injection of MMP-14 plasmid was an effective method of increasing the amount of the protein, consistent with published reports (32). Additionally, the present study showed that significantly increased MMP-14 expression existed in the standard corneal suture model and the alkali burn model. Through the examination of these models, it was shown that corneal HG and LG were significantly induced. This was in agreement with previous results showing that keratocytes and myofibroblasts express MMP-1, −2 and −9 following corneal injuries (3335). Collectively, these results demonstrate that MMP-14 is involved in corneal NV, at least under certain pathophysiological conditions. The MMP-14 overexpression in corneal tissues implied that MMP-14 plays an important role in corneal HG and LG.

Macrophages are acknowledged to have a key role in corneal LG. Previous studies have confirmed that large numbers of activated CD11b+ macrophages induce LG during corneal inflammation, by transdifferentiating into lymphatic endothelium and by releasing lymphangiogenic growth factors (36,37). As shown in the present study, the numbers of CD11b+ macrophages infiltrating the inflammatory corneas in MMP-14-treated mice were significantly greater than in vehicle-treated mice. It may be speculated that the lymphangiogenic effect of MMP-14 might also be partially caused by an indirect effect on macrophages.

To further investigate the mechanism through which MMP-14 regulates corneal HG and LG, the associations between MMP-14 and VEGF proteins and receptors were examined. A marked upregulation of VEGF-C and VEGFR-3 expression levels was detected in sutured corneas treated with MMP-14, but other members of the VEGF family exhibited no significant changes. The outgrowth of lymphatic vessels is primarily triggered by VEGF-C and its receptor VEGFR-3 (38,39), and the specific inhibition of VEGFR-3 alone is sufficient to block corneal LG (40). In previous studies, corneal LG induced by fibroblast growth factor-2 or hepatocyte growth factor could be blocked by VEGFR-3 inhibition (41,42). These investigations suggest that the VEGFR-3 signaling pathway is critical for corneal LG.

In addition, the data presented in the present study indicate that VEGF-C and its receptor VEGFR-3 might induce corneal HG in addition to LG. Among the VEGFs, VEGF-A is widely studied and has been found to be responsible for HG by binding to its receptors, VEGFR-1 and VEGFR-2 (43,44), while VEGF-C is thought to a dominant factor stimulating LG through binding to VEGFR-3. However, certain studies have provided evidence that VEGF-C is also associated with HG (4547). An earlier study reported that VEGF-C enhanced microvascular endothelial cell migration, branching and capillary sprouting in association with MMP-14 overexpression (48), which is consistent with the findings of the present study. Several studies have also shown that VEGF-C and VEGFR-3 are associated with angiogenesis in cancer (49,50). A possible mechanism by which VEGF-C induces HG has been shown to be through a RhoA-mediated pathway (51). Furthermore, it may be speculated that increased expression of VEGF-C are associated with the large numbers of activated CD11b+ macrophages in the inflammatory corneas of MMP-14-treated mice. A previous study has shown that significantly increased numbers of dermal CD11b+ macrophages expressed higher levels of VEGF-C (52), which indicates their close association. It is possible, therefore, that MMP-14 induced corneal HG and LG by upregulating the expression levels of VEGF-C and VEGFR-3 in vivo. Additional studies to elucidate the relationship between MMP-14 and VEGF-C/VEGFR-3 are necessary.

In summary, through the use of a corneal suture model, the data in the present study demonstrate that MMP-14 promotes corneal HG and LG. The important role of MMP-14 in corneal LG was closely associated with CD11b+ macrophage infiltration, and with the VEGF-C/VEGFR-3 signaling pathway. Based on these findings, MMP-14 inhibition could be a promising new target for the management of inflammatory corneal disorders and for maintaining the transparency of the cornea.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (NSFC No. 81300728), and the Scientific Research Fund of Heilongjiang Provincial Education Department (No. 12521262) and of Heilongjiang Provincial Health Bureau (2011-031).

References

1 

He Y, Rajantie I, Pajusola K, Jeltsch M, Holopainen T, Yla-Herttuala S, Harding T, Jooss K, Takahashi T and Alitalo K: Vascular endothelial cell growth factor receptor 3-mediated activation of lymphatic endothelium is crucial for tumor cell entry and spread via lymphatic vessels. Cancer Res. 65:4739–4746. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Hosseini H and Nejabat M: A potential therapeutic strategy for inhibition of corneal neovascularization with new anti-VEGF agents. Med Hypotheses. 68:799–801. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Cursiefen C, Chen L, Dana MR and Streilein JW: Corneal lymphangiogenesis: Evidence, mechanisms and implications for corneal transplant immunology. Cornea. 22:273–281. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Cursiefen C, Maruyama K, Jackson DG, Streilein JW and Kruse FE: Time course of angiogenesis and lymphangiogenesis after brief corneal inflammation. Cornea. 25:443–447. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Bachmann BO, Bock F, Wiegand SJ, Maruyama K, Dana MR, Kruse FE, Luetjen-Drecoll E and Cursiefen C: Promotion of graft survival by vascular endothelial growth factor a neutralization after high-risk corneal transplantation. Arch Ophthalmol. 126:71–77. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Cursiefen C, Cao J, Chen L, Liu Y, Maruyama K, Jackson D, Kruse FE, Wiegand SJ, Dana MR and Streilein JW: Inhibition of hemangiogenesis and lymphangiogenesis after normal-risk corneal transplantation by neutralizing VEGF promotes graft survival. Invest Ophthalmol Vis Sci. 45:2666–2673. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Lymboussaki A, Achen MG, Stacker SA and Alitalo K: Growth factors regulating lymphatic vessels. Curr Top Microbiol Immunol. 251:75–82. 2000.PubMed/NCBI

8 

Oh SJ, Jeltsch MM, Birkenhäger R, McCarthy JE, Weich HA, Christ B, Alitalo K and Wilting J: VEGF and VEGF-C: Specific induction of angiogenesis and lymphangiogenesis in the differentiated avian chorioallantoic membrane. Dev Biol. 188:96–109. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E, Diem K, Weninger W, Tschachler E, Alitalo K and Kerjaschki D: Angiosarcomas express mixed endothelial phenotypes of blood and lymphatic capillaries: Podoplanin as a specific marker for lymphatic endothelium. Am J Pathol. 154:385–394. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Alitalo K, Tammela T and Petrova TV: Lymphangiogenesis in development and human disease. Nature. 438:946–953. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova TV, Jeltsch M, Jackson DG, Talikka M, Rauvala H, et al: Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol. 5:74–80. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Patel SP and Dana R: Corneal lymphangiogenesis: Implications in immunity. Semin Ophthalmol. 24:135–138. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

14 

McCawley LJ and Matrisian LM: Matrix metalloproteinases: Multifunctional contributors to tumor progression. Mol Med Today. 6:149–156. 2000. View Article : Google Scholar : PubMed/NCBI

15 

d'Ortho MP, Will H, Atkinson S, Butler G, Messent A, Gavrilovic J, Smith B, Timpl R, Zardi L and Murphy G: Membrane-type matrix metalloproteinases 1 and 2 exhibit broad-spectrum proteolytic capacities comparable to many matrix metalloproteinases. Eur J Biochem. 250:751–757. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Koshikawa N, Giannelli G, Cirulli V, Miyazaki K and Quaranta V: Role of cell surface metalloproteinase MT1-MMP in epithelial cell migration over laminin-5. J Cell Biol. 148:615–624. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Sato H, Takino T and Miyamori H: Roles of membrane-type matrix metalloproteinase-1 in tumor invasion and metastasis. Cancer Sci. 96:212–217. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Genis L, Galvez BG, Gonzalo P and Arroyo AG: MT1-MMP: Universal or particular player in angiogenesis? Cancer Metastasis Rev. 25:77–86. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Mimura T, Chang JH, Kim TI, Onguchi T, Kojima T, Sakimoto T and Azar DT: MT1-MMP cleavage of the antiangiogenic proteoglycan decorin: Role in corneal angiogenesis. Cornea. 30:(Suppl 1). S45–S49. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Ye HQ, Maeda M, Yu FS and Azar DT: Differential expression of MT1-MMP (MMP-14) and collagenase III (MMP-13) genes in normal and wounded rat corneas. Invest Ophthalmol Vis Sci. 41:2894–2899. 2000.PubMed/NCBI

21 

Hotary K, Allen E, Punturieri A, Yana I and Weiss SJ: Regulation of cell invasion and morphogenesis in a three-dimensional type I collagen matrix by membrane-type matrix metalloproteinases 1, 2 and 3. J Cell Biol. 149:1309–1323. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C, D'Amore PA, Dana MR, Wiegand SJ and Streilein JW: VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization by macrophage recruitment. J Clin Invest. 113:1040–1050. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Stechschulte SU, Joussen AM, von Recum HA, Poulaki V, Moromizato Y, Yuan J, D'Amato RJ, Kuo C and Adamis AP: Rapid ocular angiogenic control via naked DNA delivery to cornea. Invest Ophthalmol Vis Sci. 42:1975–1979. 2001.PubMed/NCBI

24 

Cheng HC, Yeh SL, Tsao YP and Kuo PC: Subconjunctival injection of recombinant AAV-angiostatin ameliorates alkali burn induced corneal angiogenesis. Mol Vis. 13:2344–2352. 2007.PubMed/NCBI

25 

Schefe JH, Lehmann KE, Buschmann IR, Unger T and Funke-Kaiser H: Quantitative real-time RT-PCR data analysis: current concepts and the novel “gene expression's CT difference” formula. J Mol Med. 84:901–910. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Radonić A, Thulke S, Mackay IM, Landt O, Siegert W and Nitsche A: Guideline to reference gene selection for quantitative real-time PCR. Biochem Biophys Res Commun. 313:856–862. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Tang XL, Sun JF, Wang XY, Du LL and Liu P: Blocking neuropilin-2 enhances corneal allograft survival by selectively inhibiting lymphangiogenesis on vascularized beds. Mol Vis. 16:2354–2361. 2010.PubMed/NCBI

28 

Bock F, Onderka J, Hos D, Horn F, Martus P and Cursiefen C: Improved semiautomatic method for morphometry of angiogenesis and lymphangiogenesis in corneal flatmounts. Exp Eye Res. 87:462–470. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Zhang H, Hu X, Tse J, Tilahun F, Qiu M and Chen L: Spontaneous lymphatic vessel formation and regression in the murine cornea. Invest Ophthalmol Vis Sci. 52:334–338. 2010. View Article : Google Scholar

30 

Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1:27–31. 1995. View Article : Google Scholar : PubMed/NCBI

31 

Ingvarsen S, Porse A, Erpicum C, Maertens L, Jürgensen HJ, Madsen DH, Melander MC, Gårdsvoll H, Høyer-Hansen G, Noel A, et al: Targeting a single function of the multifunctional matrix metalloproteinase MT1-MMP: Impact on lymphangiogenesis. J Biol Chem. 288:10195–10204. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Onguchi T, Han KY, Chang JH and Azar DT: Membrane type-1 matrix metalloproteinase potentiates basic fibroblast growth factor-induced corneal neovascularization. Am J Pathol. 174:1564–1571. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Matsubara M, Girard MT, Kublin CL, Cintron C and Fini ME: Differential roles for two gelatinolytic enzymes of the matrix metalloproteinase family in the remodelling cornea. Dev Biol. 147:425–439. 1991. View Article : Google Scholar : PubMed/NCBI

34 

Shimoda M, Ishizaki M, Saiga T and Yamanaka N: Expression of matrix metalloproteinases and tissue inhibitor of metalloproteinase by myofibroblasts-morphological study on corneal wound healing. Nippon Ganka Gakkai Zasshi. 101:371–379. 1997.(In Japanese). PubMed/NCBI

35 

Fini ME, Girard MT and Matsubara M: Collagenolytic/gelatinolytic enzymes in corneal wound healing. Acta Ophthalmol Suppl. 26–33. 1992.PubMed/NCBI

36 

Maruyama K, Ii M, Cursiefen C, Jackson DG, Keino H, Tomita M, Van Rooijen N, Takenaka H, D'Amore PA, Stein-Streilein J, et al: Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages. J Clin Invest. 115:2363–2372. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Watari K, Nakao S, Fotovati A, Basaki Y, Hosoi F, Bereczky B, Higuchi R, Miyamoto T, Kuwano M and Ono M: Role of macrophages in inflammatory lymphangiogenesis: Enhanced production of vascular endothelial growth factor C and D through NF-kappaB activation. Biochem and Biophys Res Commun. 377:826–831. 2008. View Article : Google Scholar

38 

Ji RC: Macrophages are important mediators of either tumor- or inflammation-induced lymphangiogenesis. Cell Mol Life Sci. 69:897–914. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Su JL, Yen CJ, Chen PS, Chuang SE, Hong CC, Kuo IH, Chen HY, Hung MC and Kuo ML: The role of the VEGF-C/VEGFR-3 axis in cancer progression. Br J Cancer. 96:541–545. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Bock F, Onderka J, Dietrich T, Bachmann B, Pytowski B and Cursiefen C: Blockade of VEGFR3-signalling specifically inhibits lymphangiogenesis in inflammatory corneal neovascularisation. Graefes Arch Clin Exp Ophthalmol. 246:115–119. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Kubo H, Cao R, Brakenhielm E, Mäkinen T, Cao Y and Alitalo K: Blockade of vascular endothelial growth factor receptor-3 signaling inhibits fibroblast growth factor-2-induced lymphangiogenesis in mouse cornea. Proc Natl Acad Sci USA. 99:8868–8873. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Cao R, Björndahl MA, Gallego MI, Chen S, Religa P, Hansen AJ and Cao Y: Hepatocyte growth factor is a lymphangiogenic factor with an indirect mechanism of action. Blood. 107:3531–3536. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Shibuya M and Claesson-Welsh L: Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis. Exp Cell Res. 312:549–560. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Goodin S: Development of monoclonal antibodies for the treatment of colorectal cancer. Am J Health Syst Pharm. 65:(Suppl 4). S3–S7. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Yang ZS, Xu YF, Huang FF and Ding GF: Associations of nm23H1, VEGF-C and VEGF-3 receptor in human prostate cancer. Molecules. 19:6851–6862. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Cao R, Eriksson A, Kubo H, Alitalo K, Cao Y and Thyberg J: Comparative evaluation of FGF-2-, VEGF-A- and VEGF-C-induced angiogenesis, lymphangiogenesis, vascular fenestrations and permeability. Circ Res. 94:664–670. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Saaristo A, Veikkola T, Enholm B, Hytönen M, Arola J, Pajusola K, Turunen P, Jeltsch M, Karkkainen MJ, Kerjaschki D, et al: Adenoviral VEGF-C overexpression induces blood vessel enlargement, tortuosity and leakiness but no sprouting angiogenesis in the skin or mucous membranes. FASEB J. 16:1041–1049. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Bauer SM, Bauer RJ, Liu ZJ, Chen H, Goldstein L and Velazquez OC: Vascular endothelial growth factor-C promotes vasculogenesis, angiogenesis and collagen constriction in three-dimensional collagen gels. J Vasc Surg. 41:699–707. 2005. View Article : Google Scholar : PubMed/NCBI

49 

Laakkonen P, Waltari M, Holopainen T, Takahashi T, Pytowski B, Steiner P, Hicklin D, Persaud K, Tonra JR, Witte L and Alitalo K: Vascular endothelial growth factor receptor 3 is involved in tumor angiogenesis and growth. Cancer Res. 67:593–599. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Valtola R, Salven P, Heikkilä P, Taipale J, Joensuu H, Rehn M, Pihlajaniemi T, Weich H, de Waal R and Alitalo K: VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am J Pathol. 154:1381–1390. 1999. View Article : Google Scholar : PubMed/NCBI

51 

Kumar B, Chile SA, Ray KB, Reddy GE, Addepalli MK, Kumar AS, Ramana V and Rajagopal V: VEGF-C differentially regulates VEGF-A expression in ocular and cancer cells; promotes angiogenesis via RhoA mediated pathway. Angiogenesis. 14:371–380. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Shi VY, Bao L and Chan LS: Inflammation-driven dermal lymphangiogenesis in atopic dermatitis is associated with CD11b+ macrophage recruitment and VEGF-C up-regulation in the IL-4-transgenic mouse model. Microcirculation. 19:567–579. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 12 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Du HT and Du HT: Matrix metalloproteinase 14 participates in corneal lymphangiogenesis through the VEGF-C/VEGFR-3 signaling pathway. Exp Ther Med 12: 2120-2128, 2016
APA
Du, H., & Du, H. (2016). Matrix metalloproteinase 14 participates in corneal lymphangiogenesis through the VEGF-C/VEGFR-3 signaling pathway. Experimental and Therapeutic Medicine, 12, 2120-2128. https://doi.org/10.3892/etm.2016.3601
MLA
Du, H., Liu, P."Matrix metalloproteinase 14 participates in corneal lymphangiogenesis through the VEGF-C/VEGFR-3 signaling pathway". Experimental and Therapeutic Medicine 12.4 (2016): 2120-2128.
Chicago
Du, H., Liu, P."Matrix metalloproteinase 14 participates in corneal lymphangiogenesis through the VEGF-C/VEGFR-3 signaling pathway". Experimental and Therapeutic Medicine 12, no. 4 (2016): 2120-2128. https://doi.org/10.3892/etm.2016.3601