Please wait a minute...

中国生物工程杂志

China Biotechnology
China Biotechnology  2012, Vol. 32 Issue (02): 17-23    DOI:
    
Cloning, Expression and Analysis of Antifibrosis Function of Fusion Protein Interleukin 10 with RGD
SHI Ji-hong, SHI Shan, BAI Xiao-zhi, GUAN Hao, TAO Ke, HU Xiao-long, LIU Jia-qi, HU Da-hai
Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032,China
Download: HTML   PDF(664KB) HTML
Export: BibTeX | EndNote (RIS)      

Abstract  

Interleukin 10 (IL-10) is a pleiortropic cytokine which plays a pivotal role in the inflammatory response, immunological reaction and diseases development. RGD peptide has been demonstrated to bind to integrin on neovasculature endothelial cells specifically. A novel hybrid protein combining RGD with IL-10 was designed. The DNA sequence encoding recombination fusion protein IL10-RGD was subcloned into the pET22b(+) vector for protein expression in E. coli strain BL21(DE3). SDS-PAGE analysis showed an induced expression product band, with a molecular weight of about 19.3kDa, which was consistent with the expected value. The recombinant protein was isolated and purified by Sephadex G25M, and then renatured. Cultured human dermal fibroblasts were treated with transforming growth factor β1 (TGF-β1) stimulation and IL10-RGD treatment. The protein levels of various fibrosis related molecules were assessed by Western blot. The change of α-SMA positive fibroblasts was analyzed by immunocytochemistry. The results showed that IL10-RGD can significantly down-regulate the protein expression levels of Col1, Col3 and α-SMA, and decrease α-SMA positive cells in cultured dermal fibroblasts stimulation with TGF-β1. These findings indicate that IL10-RGD has the antifibrotic effect on dermal fibroblasts, the potential usage in fibrosis diseases by improvement the abnormal deposition of ECM, and suggest that IL10-RGD may be beneficial for the treatment of skin fibrosis and pathological scars.



Key wordsInterleukin 10      RGD peptide      Fusion expression      Antifibrosis     
Received: 24 November 2011      Published: 25 February 2012
ZTFLH:  Q789  
  Q812  
Cite this article:

SHI Ji-hong, SHI Shan, BAI Xiao-zhi, GUAN Hao, TAO Ke, HU Xiao-long, LIU Jia-qi, HU Da-hai. Cloning, Expression and Analysis of Antifibrosis Function of Fusion Protein Interleukin 10 with RGD. China Biotechnology, 2012, 32(02): 17-23.

URL:

https://manu60.magtech.com.cn/biotech/     OR     https://manu60.magtech.com.cn/biotech/Y2012/V32/I02/17


[1] Weiss E, Mamelak A J, Morgia S L, et al. The role of interleukin 10 in the pathogenesis and potential treatment of skin diseases. J Am Acad Dermatol, 2004, 50(5):657-675.

[2] Conti P, Kempuraj D, Kandere K, et al. IL-10, an inflammatory/ inhibitory cytokine, but not always. Immunol Lett, 2003, 86(2):123-129.

[3] Mocellin S, Panelli M C, Wang E, et al. The dual role of IL-10. Trends Immunol, 2003, 24(1):36-43.

[4] Occleston N L, O'Kane S, Goldspink N, et al. New therapeutics for the prevention and reduction of scarring. Drug Discovery Today, 2008, 13(21/22):973-981.

[5] Peranteau W H, Zhang L, MuvaraK N, et al. IL-10 overexpression decreases inflammatory mediators and promotes regenerative healing in an adult model of scar formation. J Invest Dermatol, 2008, 128(7):1852-1860.

[6] Assa-Munt N, Jia X, Laakkonen P, et al. Solution structures and integrin binding activities of an RGD peptide with two isomers. Biochemistry, 2001, 40(8):2373-2378.

[7] Amadeu T P, Braune A S, Mandarim-de-Lacerda C, et al. Vascularization pattern in hypertrophic scars and keloid: a stereological analysis. Pathol Res Pract, 2003, 199(7):469-473.

[8] Brown N J, Smyth E A, Cross S S, et al. Angiogenesis induction and regression in human surgical wounds. Wound Repair Regen, 2002, 10(4):245-251.

[9] Ruoslahti E. Drug targeting to specific vascular sites. Drug Discov Today, 2002, 7(22):1138-1143.

[10] Arap W, Pasqulini R, Ruoslahti E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science, 1998, 102(2):430-437.

[11] Haverstock B D. Hypertrophic scar and keloid. Clin Podiatr Med Surg, 2001, 18(1):147-159.

[12] Bloemen M C, van der Veer W M, Ulrich M M, et al. Prevention and curative management of hypertrophic scar formation. Burns, 2009, 35(4):463-475.

[13] Zhang Z F, Zhang Y G, Hu D H, et al. Smad interacting protein 1 as a regulator of skin fibrosis in pathological scars. Burns, 2011, 37(4):665-672.

[14] Sabat R, Grütz G, Warszawska K, et al. Biology of interleukin-10. Cytokine Growth Factor Rev, 2010, 21(5):331-344.

[15] Ellerby H M, Arap W, Ellerby L M, et al. Anti-cancer activity of targeted pro-apoptotic peptides. Nat Med, 1999, 5(9):1032-1038.

[16] Asadullah K, Döcke W D, Ebeling M, et al. Interleukin 10 treatment of psoriasis: clinical results of a phase 2 trial. Arch Dermatol, 1999, 135(2):187-192.

[17] Singer A J, Clark R A. Cutaneous wound healing. N Engl J Med, 1999, 341(10):738-746.

[18] Sato Y, Ohshima T, Kondo T. Regulatory role of endogenous interleukin-10 in cutaneous inflammatory response of murine wound healing. Biochem Biophys Res Commun, 1999, 265(1):194-199.

[19] Peranteau W H, Zhang L, Muvarak N, et al. IL-10 overexpression decreases inflammatory mediators and promotes regenerative healing in an adult model of scar formation. J Invest Dermatol, 2008, 128(7):1852-1860.

[20] Nakagome K, Dohi M, Okunishi K, et al. In vivo IL-10 gene delivery suppresses airway eosinophilia and hyperreactivity by down-regulating APC functions and migration without impairing the antigen-specific systemic immune response in a mouse model of allergic airway inflammation. J Immunol, 2005, 174(11):6955-6966.

[21] Hinz B. Formation and function of the myofibroblast during tissue repair. J Invest Demmatol, 2007, 127(3): 526-537.

[22] Hinz B. The myofibroblast: paradigm for a mechanically active cell. J Biometh, 2010, 43(1): 146-155.

[1] LIU Jing-ying, LIU Cheng-lang, ZHANG Yun-lei, WANG Shi-qi, XIA Li-qiu, ZHANG You-ming. The Fusion Expression of Bovine Lactoferricin (LfcinB) in Bacillus thuringiensis[J]. China Biotechnology, 2016, 36(8): 23-30.
[2] ZHU Xiao-peng, ZHANG Ya-ning, YU Jin-peng, LI Xiao-dan, LUO Su-lan, ZHANGSUN Dong-ting. Tandem Expression, Purification and Bioactivity Characterization of Conotoxin MrIA[J]. China Biotechnology, 2016, 36(5): 81-88.
[3] SHI Xian-wei, ZHANG Wei-tao, ZHANG Xiao-fei, YANG Guang-yu, FENG Yan. The Heterologous Expression and Characterization of Lytic Polysaccharide Monooxygenase from Actinosynnema mirum DSM 43827[J]. China Biotechnology, 2014, 34(7): 17-23.
[4] WANG Jian-feng, ZHANG Si-liang, WANG Yong. Pathway Assembly and Optimization in E. coli for de Novo Biosynthesis of Resveratrol[J]. China Biotechnology, 2014, 34(2): 71-77.
[5] ZHAO Hai-yang, WANG Ze, HUANG Peng-huang, TIAN Hai-shan, LI Xiao-kun, YANG Shu-lin. rhKGF2-EGFP Fusion Expression and Alcohol Liposome Preparation[J]. China Biotechnology, 2014, 34(10): 22-27.
[6] CHEN Ai-chun, PENG Wei, WANG Sheng-peng. Progress in the Application of Affinity Tags for the Expression and Purification of Recombinant Proteins[J]. China Biotechnology, 2012, 32(12): 93-103.
[7] GAO Hua, HU Chong-Bei, LI Yuan. Construction and Expression of sGLP-1- HSA Fusion protein[J]. China Biotechnology, 2009, 29(08): 19-25.
[8] . Research Advance in Recombinant Expression of Microbial Lipases[J]. China Biotechnology, 2008, 28(5): 135-140.
[9] . Expression of the Recombinant HSA-G-CSF in Pichia pastoris[J]. China Biotechnology, 2006, 26(12): 34-39.
[10] . Gene Coloning, Expression and Enzymatic Assay of Human sPLA2-IIA[J]. China Biotechnology, 2006, 26(08): 93-97.
[11] . Expression and Purification of Human Parathyroid Hormone Peptide(1-34) in Escherichia coli[J]. China Biotechnology, 2006, 26(03): 26-30.
[12] . Gene Cloning and Efficient Expression of Human Adiponectin in Escherchia coli[J]. China Biotechnology, 2006, 26(0): 15-19.