Pharmacology Study of the Multiple Angiogenesis Inhibitor RC28-E on Anti-Fibrosis in a Chemically Induced Lung Injury Model

Xiangying Kou, Yeying Sun, Shenjun Li, Weihua Bian, Zhihao Liu, Daolai Zhang, Jing Jiang, Xiangying Kou, Yeying Sun, Shenjun Li, Weihua Bian, Zhihao Liu, Daolai Zhang, Jing Jiang

Abstract

Background: Disease-related injury in any organ triggers a complex cascade of cellular and molecular responses that culminate in tissue fibrosis, inflammation, and angiogenesis simultaneously. Multiple cell angiogenesis is an essential part of the tissue damage response, which is involved in fibrosis development. RC28-E is a novel recombinant dual decoy receptor lgG1 Fc-fusion protein that can block vascular endothelial growth factor (VEGFA), platelet-derived growth factor (PDGF), and fibroblast growth factor-2 (FGF-2) simultaneously. This protein has stepped into clinical trials (NCT03777254) for the treatment of pathological neovascularization-related diseases. Here, we report on the role of RC28-E during anti-fibrosis and its potential multitarget function in regulating fibrosis.

Methods: A bleomycin-induced pulmonary fibrosis C57BL/6 mouse model was established. Hematoxylin and eosin staining (HE) and Masson staining (Masson's) were performed to evaluate the pulmonary fibrosis based on the scoring from, Ashcroft score. Fibrosis related factors and inflammatory cytokines including HYP, α-SMA, procollagen, ICAM, IL-6, IL-1, and TNF-α were also determined at the protein and mRNA levels to characterize the fibrosis. Both mRNA and protein levels of VEGF, FGF, and transforming growth factor (TGF)-β were detected by quantitative real-time PCR (qRT-PCR) and immunohistochemical (IHC) analysis, respectively. Pulmonary fibrosis and related cytokines were re-evaluated in vivo after 3 doses of RC28-E (5 mg/kg, 15 mg/kg, and 50 mg/kg, ip. Tiw × 9) in comparison with a mono-target antagonist treatment (VEGF or FGF blocking). RC28-E attenuated the activation of TGF-β induced fibroblasts in vitro. Expression levels of α-SMA and collagen I, as well as proliferation and migration, were determined with the human skin fibroblast cell line Detroit 551 and primary murine pulmonary fibroblast cells. The mechanism of RC28-E via the TGF-β/Smad pathway was also investigated.

Results: RC28-E exhibits significant anti-fibrosis effects on Idiopathic pulmonary fibrosis (IPF) in vivo. Moreover, TGF-β induced fibroblast activation in vitro via the inhibition of the TGF-β downstream Smad pathway, thus providing potential therapeutics for clinical disease-related fibrosis-like IPF as well as chemotherapy-induced fibrosis in cancer therapy.

Keywords: FGF; VEGF; angiogenesis; anti-fibrosis; lung injury; pharmacology study.

Conflict of interest statement

The authors declare that they have no conflict of interest concerning this article.

Figures

Figure 1
Figure 1
Schematic diagram of RC28 molecular design. VEGFR1: vascular endothelial growth factor receptor 1; FGFR1: fibroblast growth factor receptor 1.
Figure 2
Figure 2
Pathological changes in lung tissue and the level of inflammatory cytokine in model of mice after bleomycin-induced pulmonary fibrosis formation. (A) The lung section from the mice that have undergone different treatments were stained by hematoxylin–eosin (HE) (n = 4 per group). (B) Lung sections from various treatment groups were subjected to Masson trichrome staining (n = 4 per group). (C) The expression of α-SMA was determined by using immunohistochemical analysis. Scale bar = 200 μm for each picture (original magnification: ×100, n = 4 per group). (D) The lung fibrosis score based on the scoring from Ashcroft score. (E) Quantitative analysis of α-SMA positive area. (F) Hydroxyproline concentration in lung tissue was detected by using alkaline hydrolysis (n = 4 per group). (G) Change of α-SMA, procollagen, and ICAM transcripts were analyzed by qRT-PCR (n = 6 per group). (H) The expression of IL-6, IL-1, and TNF-α was examined by using ELISA in the lung and serum. All data are expressed as the mean ± SD. #p < 0.05, ##p < 0.01 versus control.
Figure 3
Figure 3
Expression levels of VEGF, FGF, and TGF-β in lung of mice after bleomycin-induced pulmonary fibrosis formation. (AC) The VEGF, FGF, and TGF-β expression levels in the control mice and modeling mice were determined by immunohistochemistry assay. Scale bar = 200 μm for each picture (original magnification: X100, n = 4 per group). Quantitative analysis of VEGF, FGF, and TGF-β positive area. Densitometry data are shown as mean ± SD. (D) The levels of VEGF, FGF, and TGF-β mRNA in lung tissue were detected by qRT-PCR (n = 6 per group). Results are shown as mean ± SD. #p < 0.05, ##p < 0.01 versus control.
Figure 4
Figure 4
Effect of RC28-E on VEGF, FGF, and TGF-β expression in bleomycin-induced fibrosis formation. (AC) Immunohistochemistry was used to examine VEGF, FGF, and TGF-β expression in lung tissues. Scale bar = 200 μm for each picture (original magnification: ×100). (DF) The change of VEGF, FGF, and TGF-β transcripts were analyzed by qRT-PCR. (n = 4 per group). Results are shown as mean ± SD. * p < 0.05, ** p < 0.01 versus model alone. #p < 0.05, ##p < 0.01 versus control.
Figure 5
Figure 5
The effect of RC28-E on the development of fibrosis in a therapeutic mouse bleomycin model. (A) The lung sections from the mice that have undergone different treatments were stained by hematoxylin–eosin (HE). (B) Lung sections from various treatment groups were subjected to Masson trichrome staining. (C) Immunohistochemistry was used to examine α-SMA expression in pulmonary tissues. Scale bar = 200 μm for each picture (original magnification: ×100, n = 4 per group). (D) The lung fibrosis score based on the scoring from Ashcroft score. (E) Quantitative analysis of α-SMA positive area. (F) The effect of RC28-E on hydroxyproline concentration in lung tissue. (GI) The expression of α-SMA, procollagen, and ICAM mRNA were detected by qRT-PCR. (JL) IL-1, IL-6, and TNF-α ELISA kit were used to detect the IL-1, IL-6, and TNF-α expression in the bleomycin induced lung tissue. Results are shown as mean ± SD. * p < 0.05, ** p < 0.01 versus model alone. #p < 0.05, ##p < 0.01 versus control.
Figure 6
Figure 6
RC28-E attenuates TGF-β-induced fibroblast activation. (AD) Human skin fibroblasts line Detroit 551 were treated with RC28-E (2400 nM) or 2400 nM RC28-C1 (VEGF-trap) and RC28-C2 (FGF-trap) in the presence hTGF-β1 (10 ng/mL) for 72 h. (A) The protein expression levels of α-SMA and collagen I were examined by western blot and GAPDH as loading control. Quantitative data were from western blot analysis (n = 3). (B) The expression of α-SMA was detected by immunofluorescence assay. Nuclei were stained with DAPI (blue). Scale bar = 275 μm for each picture (original magnification: ×200). (C,D) Changes of α-SMA and procollagen transcripts were analyzed by qRT-PCR. (E) cells were treated with 1200, 1600, 2000, and 2400 nM RC28-E or 1200, 1600, 2000, and 2400 nM RC28-C1 (VEGF-trap) or 1200, 1600, 2000, and 2400 nM RC28-C2 (FGF-trap), with hTGF-β1 (10 ng/mL) as the control, for 48 h. Relative cell proliferation was determined by using cell counting kit-8. (F) Transwell was used to migration analysis, the migrated cells were stained with crystal violet. Results are shown as mean ± SD. * p < 0.05, ** p < 0.01 versus hTGF-β1 alone. #p < 0.05, ##p < 0.05 versus control (n = 3).
Figure 7
Figure 7
RC28-E downregulated protein levels of VEGF and FGF and inhibited the phosphorylation activity of the downstream signaling molecules Smad in human skin fibroblasts. (AE) Human skin fibroblasts Detroit 551 were pretreated with RC28-E (2400 nM) for 1 h, followed by the addition of hTGF-β1 into the culture medium to a final concentration of 10 ng/mL and incubation for 72 h. (A,B) The protein levels of VEGF and FGF were examined by ELISA. (C,D) The expression of VEGF and FGF mRNA were analyzed by qRT-PCR. (E) The expression of P-smad2, P-smad3, and smad2/3 was determined using western blot and GAPDH as loading control. Representative gel electrophoresis bands are shown, and the expression levels of proteins were quantified by densitometry and normalized to the expression of smad2/3. Densitometry data are shown as mean ± SD. * p < 0.05, ** p < 0.01 versus hTGF-β1 alone. #p < 0.05, ##p < 0.05 versus control (n = 3).

References

    1. Zeisberg M., Kalluri R. Cellular mechanisms of tissue fibrosis: 1. common and organ-specific mechanisms associated with tissue fibrosis. Am. J. Physiol. Cell Physiol. 2013;304:C216–C225. doi: 10.1152/ajpcell.00328.2012.
    1. Salem C., Slim R., Fathallah N. Fibrosis—A common pathway to organ injury and failure. New Engl. J. Med. 2015;373:95–96.
    1. Liu Y. Cellular and molecular mechanisms of renal fibrosis. Nat. Rev. Nephrol. 2011;7:684–696. doi: 10.1038/nrneph.2011.149.
    1. Kaissling B., Lehir M., Kriz W. Renal epithelial injury and fibrosis. Biochim. Biophys. Acta. 2013;1832:931–939. doi: 10.1016/j.bbadis.2013.02.010.
    1. Chen J., Chen J.-K., Nagai K., Plieth D., Tan M., Lee T.C., Threadgill D.W., Neilson E.G., Harris R.C. EGFR signaling promotes TGFβ-dependent renal fibrosis. J. Am. Soc. Nephrol. 2012;23:215–224. doi: 10.1681/ASN.2011070645.
    1. Mezzano S.A., Ruiz-Ortega M., Egido J. Angiotensin II and renal fibrosis. Hypertension. 2001;38:635–638. doi: 10.1161/hy09t1.094234.
    1. Saylor P.J., Escudier B., Michaelson M.D. Importance of fibroblast growth factor receptor in neovascularization and tumor escape from antiangiogenic therapy. Clin. Genitourin. Cancer. 2012;10:77–83. doi: 10.1016/j.clgc.2012.01.010.
    1. Sun X.T., Ding Y.T., Yan X.G., Wu L.Y., Li Q., Cheng N., Qiu Y.D., Zhang M.Y. Angiogenic synergistic effect of basic fibroblast growth factor and vascular endothelial growth factor in an in vitro quantitative microcarrier-based three-dimensional fibrin angiogenesis system. World J. Gastroenterol. 2004;10:2524–2528. doi: 10.3748/wjg.v10.i17.2524.
    1. Giavazzi R., Sennino B., Coltrini D., Garofalo A., Dossi R., Ronca R., Tosatti M.P.M., Presta M. Distinct role of fibroblast growth factor-2 and vascular endothelial growth factor on tumor growth and angiogenesis. Am. J. Pathol. 2003;162:1913–1926. doi: 10.1016/S0002-9440(10)64325-8.
    1. Jhappan C., Stahle C., Harkins R.N., Fausto N., Smith G.H., Merlino G.T. TGFcalpha overexpression in transgenic mice induces liver neoplasia and abnormal development of the mammary gland and pancreas. Cell. 1990;61:1137–1146. doi: 10.1016/0092-8674(90)90076-Q.
    1. Presta M., Dell’Era P., Mitola S., Moroni E., Ronca R., Rusnati M. Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 2005;16:159–178. doi: 10.1016/j.cytogfr.2005.01.004.
    1. Compagni A., Wilgenbus P., Impagnatiello M.A., Cotton M., Christofori G. Fibroblast growth factors are required for efficient tumor angiogenesis. Cancer Res. 2000;60:7163–7169.
    1. Ogata T., Miyauchi T., Irukayama-Tomobe Y., Takanashi M., Goto K., Yamaguchi I. The peroxisome proliferatoractivated receptor alpha activator fenofibrate inhibits endothelin-1-induced cardiac fibroblast proliferation. J. Cardiovasc Pharm. 2004;44:S279–S282. doi: 10.1097/01.fjc.0000166274.24797.0e.
    1. Strhutz F., Zeisberg M., Ziyadeh F.N., Yang C.Q., Kalluri R., Müller G.A., Neilson E.G., Renziehausen A., Sisic Z. Role of basic fibroblast growt factor-2 in epithelial-mesenchymal transformation. Kidney Int. 2002;61:1714–1728. doi: 10.1046/j.1523-1755.2002.00333.x.
    1. Yu C., Wang F., Jin C., Huang X., Miller D.L., Basilico C., McKeehan W.L. Role of fibroblast growth factor type 1 and 2 in carbon tetrachloride-induced hepatic injury and fibrogenesis. Am. J. Pathol. 2003;163:1653–1662. doi: 10.1016/S0002-9440(10)63522-5.
    1. Triantafylla M., Massa H.F., Dardabounis D., Gatzioufas Z., Kozobolis V., Ioannakis K., Perente I., Panos G.D. Ranibizumab for the treatment of degenerative ocular conditions. Clin. Ophthalmol. 2014;8:1187–1198.
    1. Blick S.K., Keating G.M., Wagstaff A.J. Ranibizumab. Drugs. 2007;67:1199–1206. doi: 10.2165/00003495-200767080-00007.
    1. Yilmaz A., Karatay M., Yildirim T., Celik H., Sertbas I., Erdem Y., Caydere M., Isi H.S., Bayar M.A. Prevention of epidural fibrosis using ranibizumab in a postlaminectomy rat model. Turk. Neurosurg. 2017;27:119–123. doi: 10.5137/1019-5149.JTN.14882-15.1.
    1. Denizli N., Demirci R., Duranay M., Yilmaz B. New insight into treatment of diseases of glomerular endothelium. Turk. Nephrol. Dial. Transplant. J. 2011;20:220–226. doi: 10.5262/tndt.2011.1003.03.
    1. Huang Y., Feng H., Kan T., Huang B., Zhang M., Li Y., Shi C., Wu M., Luo Y., Yang J., et al. Bevacizumab attenuates hepatic fibrosis in rats by inhibiting activation of hepatic stellate cells. PLoS ONE. 2013;8:e73492. doi: 10.1371/journal.pone.0073492.
    1. Kahook M.Y. Bleb morphology and vascularity after trabecu-lectomy with intravitreal ranibizumab: A pilot study. Am. J. Ophthalmol. 2010;150:399–403. doi: 10.1016/j.ajo.2010.03.025.
    1. Nilforushan N., Yadgari M., Kish S.K., Nassiri N. Subcon-junctival bevacizumab versus mitomycin C adjunctive to trabeculectomy. Am. J. Ophthalmol. 2012;153:352–357. doi: 10.1016/j.ajo.2011.08.005.
    1. Grimminger F., Gunther A., Vancheri C. The role of tyrosine kinases in the pathogenesis of idiopathic pulmonary fibrosis. Eur. Respir. J. 2015;45:1426–1433. doi: 10.1183/09031936.00149614.
    1. Park H.-Y.L., Kim J.H., Park C.K. VEGF induces TGF-β1 expres-sion and myofibroblast transformation after glaucoma surgery. Am. J. Pathol. 2013;182:2147–2154. doi: 10.1016/j.ajpath.2013.02.009.
    1. Holderfield M.T. Crosstalk between vascular endothelial growth factor, notch, and transforming growth factor-beta in vascular morphogenesis. Circ. Res. 2008;102:637–652. doi: 10.1161/CIRCRESAHA.107.167171.
    1. Barratt S.L., Flower V.A., Pauling J.D. VEGF (vascular endothelial growth factor) and fibrotic lung disease. Int. J. Mol. Sci. 2018;19:1269. doi: 10.3390/ijms19051269.
    1. Kinoshita T. Molecular mechanisms of pulmonary fibrogenesis and its progression to lung cancer: A review. Int. J. Mol. Sci. 2019;20:1461. doi: 10.3390/ijms20061461.
    1. Jiang J., Xu K., Wang L., Xin W., Zhao G., Huang M., Li S., Luan X., Fang J. Pharmacology study of a chimeric decoy receptor trap fusion protein on retina neovascularization by dual blockage of VEGF and FGF-2. Eur. J. Pharm. Sci. 2018;121:251–259. doi: 10.1016/j.ejps.2018.04.043.
    1. Li D., Xie K., Zhang L., Yao X., Li H., Xu Q., Wang X., Jiang J., Fang J. Dual blockade of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (FGF-2) exhibits potent anti-angiogenic effects. Cancer Lett. 2016;377:164–173. doi: 10.1016/j.canlet.2016.04.036.
    1. Yang Q.H., Zhang Y., Jiang J., Wu M.M., Han Q., Bo Q.Y., Yu G.-W., Ru Y.S., Liu X., Huang M., et al. Protective effects of a novel drug RC28-E blocking both VEGF and FGF2 on early diabetic rat retina. Int. J. Ophthalmol. 2018;11:935–944. doi: 10.18240/ijo.2018.06.07.
    1. Ashcroft T., Simpson J.M., Timbrell V. Simple method of estimating severity of pulmonary fibrosis on a numerical scale. J. Clin. Pathol. 1998;41:467–470. doi: 10.1136/jcp.41.4.467.
    1. Tao H., Yang J.J., Shi K.H. Wnt signaling pathway in cardiac fibrosis: New insights and directions. Metabolism. 2016;65:30–40. doi: 10.1016/j.metabol.2015.10.013.
    1. Ferrari G., Pintucci G., Seghezzi G., Hyman K., Galloway A.C., Mignatti P. VEGF, a prosurvival factor, acts in concert with TGF-1 to induce endothelial cell apoptosis. Proc. Natl. Acad. Sci. USA. 2006;103:17260–17265. doi: 10.1073/pnas.0605556103.
    1. Mishra L., Derynck R., Mishra B. Transforming growth factor-beta signaling in stem cells and cancer. Science. 2005;310:68–71. doi: 10.1126/science.1118389.
    1. Rahimi R.A., Leof E.B. TGF-beta signaling: A tale of two responses. J. Cell Biochem. 2007;102:593–608. doi: 10.1002/jcb.21501.
    1. Hoshino M., Takahashi M., Aoike N. Expression of vascular endothelial growth factor, basic fibroblast growth factor, and angiogenin immunoreactivity in asthmatic airways and its relationship to angiogenesis. J. Allergy Clin. Immunol. 2001;107:295–301. doi: 10.1067/mai.2001.111928.
    1. Hamada N., Kuwano K., Yamada M., Hagimoto N., Hiasa K., Egashira K., Nakashima N., Maeyama T., Yoshimi M., Nakanishi Y. Anti-vascular endothelial growth factor gene therapy attenuates lung injury andfibrosis in mice. J. Immunol. 2005;175:1224–1231. doi: 10.4049/jimmunol.175.2.1224.
    1. Kim K.K., Sheppard D., Chapman H.A. TGF-β1 signaling and tissue fibrosis. Cold Spring Harb. Perspect. Biol. 2018;10 doi: 10.1101/cshperspect.a022293.
    1. Wang X., Ma W., Han S., Meng Z., Zhao L., Yin Y., Wang Y., Li J. TGF-β participates choroid neovascularization through Smad2/3-VEGF/TNF-α signaling in mice with Laser-induced wet age-related macular degeneration. Sci. Rep. 2017;7:9672. doi: 10.1038/s41598-017-10124-4.
    1. Mishra A., Mishra S.C., Tripathi A.M. Clinical correlation of molecular (VEGF, FGF, PDGF, c-Myc, c-Kit, Ras, p53) expression in juvenile nasopharyngeal angiofibroma. Eur. Arch. Oto-Rhino-L. 2018;275:2719–2726. doi: 10.1007/s00405-018-5110-5.
    1. Tang B., Vu M., Booker T., Santner S.J., Miller F.R., Anver M.R., Wakefield L.M. TGF-beta switches fromtumor suppressor to prometastatic factor in a model of breast cancer progression. J. Clin. Investig. 2003;112:1116–1124. doi: 10.1172/JCI200318899.
    1. Robertson I.B., Horiguchi M., Zilberberg L., Dabovic B., Hadjiolova K., Rifkin D.B. Latent TGF-β-binding proteins. Matrix. Biol. 2015;47:44–53. doi: 10.1016/j.matbio.2015.05.005.
    1. Huang X.R., Chung A.C.K., Zhou L., Wang X.J., Lan H.Y. Latent TGF-β1 protects against crescentic glornerulonephritis. J. Am. Soc. Nephrol. 2008;19:233–242. doi: 10.1681/ASN.2007040484.
    1. Xu F.Y., Liu C.W., Zhou D.D., Zhang L. TGF-/SMAD pathway and its regulation in hepatic fibrosis. J. Histochem. Cytochem. 2016;64:157–167. doi: 10.1369/0022155415627681.
    1. Zhao J.S., Shi W., Wang Y.L., Chen H., Bringas P., Datto M.B., Frederick J.P., Wang X.F., Warburton D. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am. J. Physiol.-Lung C. 2002;282:L585–L593. doi: 10.1152/ajplung.00151.2001.
    1. Tang P.M., Zhou S., Li C.J., Liao J., Xiao J., Wang Q.M., Lian G.Y., Li J., Huang X.R., To K.F., et al. The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring. Kidney Int. 2018;93:173–187. doi: 10.1016/j.kint.2017.07.026.
    1. Andrianifahanana M., Wilkes M.C., Gupta S.K., Rahimi R.A., Repellin C.E., Edens M., Wittenberger J., Yin X., Maidl E., Becker J., et al. Profibrotic TGFβ responses require the cooperative action of PDGF and ErbB receptor tyrosine kinases. FASEB J. 2013;27:4444–4454. doi: 10.1096/fj.12-224907.
    1. Li C.M., Khosla J., Pagan I., Hoyle P., Sannes P.L. TGF-beta1 and fibroblast growth factor-1 modify fibroblast growth factor-2 production in type II cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2000;279:L1038–L1046. doi: 10.1152/ajplung.2000.279.6.L1038.
    1. Kucich U., Rosenbloom J.C., Herrick D.J., Abrams W.R., Hamilton A.D., Sebti S.M., Rosenbloom J. Signaling events required for transforming growth factor-beta stimulation of connective tissue growth factor expression by cultured human lung fibroblasts. Arch. Biochem. Biophys. 2001;395:103–112. doi: 10.1006/abbi.2001.2571.
    1. Cantley L.C. The phosphoinositide 3-kinase pathway. Science. 2002;296:1655–1657. doi: 10.1126/science.296.5573.1655.
    1. Roper J., Richardson M.P., Wang W.V., Richard L.G., Chen W., Coffee E.M., Sinnamon M.J., Lee L., Chen P.C., Bronson R.T., et al. The dual PI3K/mTOR inhibitor NVP-BEZ235 induces tumor regression in a genetically engineered mouse model of PIK3CA wild-type colorectal cancer. PLoS ONE. 2011;6 doi: 10.1371/journal.pone.0025132.
    1. Turner N., Grose R. Fibroblast growth factor signalling: From development to cancer. Nat. Rev. Cancer. 2010;10:116–129. doi: 10.1038/nrc2780.

Source: PubMed

3
Se inscrever