?

Recombinant expression and purification of functional vascular endothelial growth factor-121 in the baculovirus expression system

2017-01-19 07:35NastaranMohseniAliJahanianNajafabadiFatemeKazemiLomedashtRoghayeArezomandMahdiHabibiAnbouhiDelavarShahbazzadehMahdiBehdani
關鍵詞:產品品質低端兵團

Nastaran Mohseni, Ali Jahanian Najafabadi, Fateme Kazemi-Lomedasht, Roghaye Arezomand, Mahdi Habibi-Anbouhi, Delavar Shahbazzadeh, Mahdi Behdani?

1Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran2Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran3Department of Medical Biotechnology and Molecular Science, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran4National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran

Recombinant expression and purification of functional vascular endothelial growth factor-121 in the baculovirus expression system

Nastaran Mohseni1, Ali Jahanian Najafabadi2, Fateme Kazemi-Lomedasht1, Roghaye Arezomand3, Mahdi Habibi-Anbouhi4, Delavar Shahbazzadeh1, Mahdi Behdani1?

1Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran
2Department of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
3Department of Medical Biotechnology and Molecular Science, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran
4National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran

ARTICLE INFO

Article history:

Received in revised form 26 August 2016

Accepted 5 September 2016

Available online 20 December 2016

Vascular endothelial growth factor Baculovirus expression system

Objective:To express human Vascular endothelial growth factor121 (VEGF121) in insect cells.Methods: A gene construct containing VEGF was cloned in the pFastBac-HTA vector, followed by transformation in DH10BAC. The recombinant bacmid was then extracted, and transfected into Sf9 insect cells. The transfected cells were harvested, and then VEGF expression was confirmed by Western blotting using specific antibodies. The tube formation assay was used for functional assessment of VEGF.Results: Our results showed that VEGF could be successfully expressed in the baculovirus system. Purified VEGF was able to stimulate in vitro tube formation of human endothelial cells.Conclusions: Results from this study demonstrated that the recombinantly-produced VEGF can be considered as a promising candidate for therapeutic purposes.

1. Introduction

Angiogenesis refers to the formation and development of new blood vessels from pre-existing blood vessels [1]. Angiogenesis is a rare phenomenon in healthy adult, which only occur temporarily under specific physiological conditions such as wound healing, inflammation and women's sexual cycle [2, 3]. A disruption in the balance between angiogenic and anti-angiogenic factors leads to pathological angiogenesis [4-6]. There is strong evidence that tumor cells need new blood vessels to grow, invasion and spread[7, 8]; in the absence of blood vessels, tumor cells can not exceed more than 1-2 mm in size [9]. Vascular endothelial growth factor (VEGF) is the most important angiogenic factor [4, 10-12], which includes 5 members in mammals such as VEGF-A, VEGF-B, VEGF-C, VEGF-D and PlGE (Placental growth factor). VEGF-A, the most important member of this family [13], interacts with two receptors, VEGFR-2 and VEGFR-1, resulting in endothelial cell proliferation and angiogenesis [14, 15]. VEGF-A consists of 5 main isoforms with different amino acid residues, including VEGF121,VEGF145,VEGF165,VEGF189 and VEGF206. Improtantly, VEGF121 and VEGF165 are the most abundant isoforms with 121 and 165 amino acid residues, respectively [16-18]. These isoforms are found in the majority of cells expressing the VEGF gene. The presence of exon 7 in the VEGF165 gene, but not in the VEGF121 gene, enables VEGF165 to bind to heparin, heparan sulfate and neuropilin-1(NP-1) receptor, which play a role in the development of embryo [16]. The low molecular mass of VEGF121 make it soluble and freely diffusible [19]. A variety of studies demonstrated that VEGF121 displays full biological activities of larger isoforms [19], emphasizing the role of VEGF121 as apotential target for drug development. Difficulties in the production of recombinant proteins, such as VEGF, inEscherichia coli(E. coli) lead to the development of novel expression systems such as the baculovirus expression system [20]. Insect cells are demonstrated to be an excellent alternative for the production of animal proteins [21]. By definition, insect cell expression systems that use insect viruses are considered as a baculoviruse. Simply stated, baculoviruses are a group of viruses that infect vertebrates and insects [22]. One of the important advantages of these systems over bacterial expression systems is to mediate post-translational modifications such as phosphorylation, glycosylation and acylation. The formation and reduction of disulfide bonds (proteolytic characteristics) result in the production of recombinant proteins, which is structurally similar to natural proteins. Another important benefit of this system is to allow low-cost production of recombinant proteins [23]. In this regard, the aim of this study was to express VEGF in the baculoviruse system.

2. Material and methods

2.1. VEGF121 DNA construct preparation

The human VEGF121 gene was cloned in pET26b in our previous study [24]. In the present study, we attampted to clone hVEGF into the pFastBac HTA transfer plasmid (Bac-to-Bac expression system). For this purpose, the hVEGF gene was amplified with specific primers anchored with BamHI and XhoI restriction enzyme sites (Table 1). Amplified hVEGF was cloned in the pFastBac HTA vector. Subsequently, the construct was transformed into E. coli TG1 competent cells, and confirmed by colony-PCR with POLH-F and PFSBC-R primers (Table 1). The fidelity of cloned sequences was verified by DNA sequencing; the resulting construct was named pFast-hVEGF (Figure 1A).

Table 1 Primers sequence.

2.2. Transposition of the VEGF121 gene to baculovirus Bacmid

該研究分析了兵團農機發展現狀與發展趨勢,根據統計數據分析,兵團農業機械化需求還有較大的空間。以犁具生產技術為例,分析了兵團犁具應用情況、兵團犁具技術研究情況、兵團犁具生產企業現狀,兵團當前存在農機生產企業管理水平低,產品品質低端,科研投入不足,產品低水平仿制嚴重,市場競爭惡化,人才欠缺、流失嚴重,新技術、新產品轉化推廣程度低等一系列問題,這些問題凸顯了兵團農機行業改革的必要性與緊迫性。建議從企業管理水平、開發力度、 機具適應性、質量監督、培訓推廣、產品售后等方面著手解 決,進行資源整合去低端產能,從而提高產品品質與經濟 效益。

The pFast-hVEGF construct was transformed into DH10Bac bacteria to establish recombinant baculovirus bacmid (Bac-to-Bac baculovirus expression system). In the Bac-to-Bac baculovirus expression system, transposition generally occurs between two areas of mini-Tn7 pFastBac and mini-att Tn7 baculovirus Bacmid. Replacement of mini-Tn7 at the junction of Bacmid results in LacZ disruption. Therefore, the recombinant Bacmid was screened after 48 h using blue-white colony screening on LB agar containing 50 μg/ μL kanamycin, 25 μg/μL tetracycline and 40 μg/μL gentamicin, and incubated at 37°℃ for 18 h. Fidelity of the VEGF gene transferred into Bacmid was confirmed by PCR using M13 primers (Table 1). The recombinant bacmid was extracted from DH10Bac according to the standard method [25], and used for transfection of Sf9 insect cells.

2.3. Sf9 Culture and transfection

Sf9 cells (obtained from National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran) were cultured in Grace’s insect medium (Invitrogen, gibco) supplemented with 10% FBS, 100 U/ mL of penicillin, 100 mg/mL of streptomycin, and incubated at 27 ℃ in a humidified incubator in an atmosphere of 5% CO2. The recombinant bacmid construct was transfected to exponentiallygrowing Sf9 cells using a cellfectin (Invitrogen) transfection reagent according to the manufacturer’s instructions.

2.4. Recombinant baculovirus preparation

Cytopathic effects (CPEs) were detected 4 days after transfection. The supernatant of Sf9 cells was collected (P1 or First Generation of virus), and used to infect new Sf9 cells. The P1 virus was used for Sf9 transfection to establish P2 and P3. To confirme VEGF expression, cells transfected by P1, P2 and P3 were harvested after four days of infection. Then, expression of recombinant VEGF121 was evaluated by SDS-PAGE and western blotting on cell lysates. Avastin (Commercial Anti-VEGF, at a dilution of 1:4000) and HRP-conjugated Anti-Human IgG (at a dilution of 1:4000) were used as primary and secondary antibodies in western blot analysis, respectively.

2.5. Purification of the recombinant protein

For VEGF purification, a density of approximately 60×106Sf9 transfected cells were harvested, and then suspended in 5 ml PBS. The cells were disrupted by three freeze/thaw cycles, and subsequently suspended in lysis buffer (50 mM NaH2PO4, 300 mM NaCl, 10 m Mimidazole), followed by 15 ultrasonication treatments of 15 sec each. The mixture was centrifuged at 6 000 g for 30 min at4 ℃ to remove insoluble particles. Supernatants were filtered using 0.45 μm syringe filters, and then loaded on the nickel affinity column (QIAGEN, Germany). The column was washed using washing buffer (50 mM NaH2PO4, 300 mM NaCl, 20 m Mimidazole) to elute unbound proteins. The recombinant His-tagged VEGF was eluted using elution buffer (50 mM NaH2PO4, 300 mM NaCl, and 250 mM imidazole). The purification process was confirmed by 15% SDS-PAGE and western blot analysis. Expression levels were detected by the Bradford assay.

2.6. Tube assay

For tube formation assay, we used primary HUVECs (Human Umbilical Vein Endothelial Cells) between passages 2 and 6, as isolated in our previous study [26]. A solution of GeltrexTM LDEV-Free Reduced Growth Factor Basement Membrane (Gibco, Invitrogen) was thawed at 4 ℃, coated at a dose of 50 μL on a 96-well plate, and incubated at 37 ℃ in a humanified incubator for 30 min in order to solidify the Geltrex. 100 μL of EBM-2 medium containing 5 000 HUVEC cells with and without of recombinant expressed VEGF was added to each well. After 2, 4 and 6 hours, cell conditions and tube formation were monitored under an invert microscope (INV100-FL, BEL-Italy). In addition, it is important to note that the assay was carried out in triplicate.

3. Results

3.1. hVEGF121 amplification and recombinant baculovirus DNA preparation

As shown in Figure 1B, the amplified human VEGF121 (400 bp) was detected in gel electrophoresis. hVEGF121 and pfastBac were digested with BamHI and XhoI, ligated and transformed into E. coli TG1 competent cells. Approximately 24 colonies were screend by colony-pcr, 21 colonies of which contained recombinant hVEGF121 (Figure 1C). In addition, BamHIand XhoI double digestion (Figure 1D) and sequencing confirmed the fidelity of recombinant hVEGF121. Finally, recombinant pfast-hVEGF121 was transformed into DH10Bac, and recombinant baculoviral DNA resulted from the Bac-to-Bac baculovirus expression system. Results from PCR, carried out on baculovirs plasmid, showed that recombinant baculovirus yielded a band of 2740bp (Figure 1 E and 2 E).

3.2. Transfection of Sf9 cells

Transfection of Sf9 cells was confirmed by the recombinant baculovirus (Figure 2). After 72-h post transfection, cytopathic effects (CPEs) were found in transfected Sf9 cells as compared to non-transfected Sf9cells (control cells), indicating the successful Transfection (Figure 3A). The cells transfected with recombinant baculovirus showed CPEs of nucleus enlargement and granulation (Figure 3B).

3.3. Expression and purification of recombinant hVEGF

After 3 days of Transfection when CPEs were observed, Sf9 cells were harvested, and expression of h VEGF121 was evaluated by using 15% SDS-PAGE and western blot analysis. As shown in Figure 4A, h VEGF121 was expressed in transfected cells; two protein bands (15 to 40 kDa) indicate different glycosylated forms of the protein. For h VEGF121 purification, 60×106 of Sf9 cells were disrupted by sonication, and cell debris was removed by centrifugation. The supernatant was loaded on the Ni-NTA column, and recombinant protein was eluted by imidazol 500mM. The yield ofpurified recombinant VEGF121 per liter of Sf9 cells was about 500 μg.

Figure 1. A) Schematic structure of the VEGF gene in the plasmid pFastBac HTa. B)Amplified hVEGF121 (400bp). C) Colony-PCR results (after cloning of hVEGF121 in pFastBac HTA plasmid). D) Double digestion of pFast-hVEGF resulting plasmid with BamHI and XhoI. Lane 1, undigested and lane 2, double digested. E) DNA electerophoresis of isolated recombinant bacmid (lanes 1, 2) and non-recombinant bacmid (lane 3) with lambda phage marker.

Figure 2. Blue-white colony screening for transformation of pFastBac-VEGF in DH10Bac.

Figure 3. Transfection of Sf9 cells by recombinant bacmid DNA.

Figure 4. A) SDS-PAGE results, 1; non-transfected cells, 2; transfected cells.

3.4. Tube formation assay

The findings from this study indicated that recombinant VEGF121 has the ability to increase tube formation in HUVECs. As shown in Figure 5, VEGF121-treated HUVECs were able to form tube-like structures. By contrast, there were no tube like structures in control cells (HUVECs with no VEGF). The cells were stained with calcein AM (Trevigen) according to the manufacturer’s protocols.

Figure 5. Tube assay resalt. A) the lack of VEGF. B) the presence of VEGF.

4. Discussion

Nowadays, there is a worldwide demand for recombinant protein production, as one of the main interest of pharmaceutical industries[27]. The most important decisions in the selection of recombinant protein production and purification are related to the host that affect product quality. To choose an appropriate host, some important criteria must be considered, including the amount of product required, toxicity, ease of purification and biological activity of the product [28]. There had been several expression systems, each with its own advantages and disadvantages. Today, the E.coli expression system is the most commonly used expression system for recombinant protein production. The best-known benefits for this system are the ability to grow fast, simple and cheap requirements, high-density environments, well-known genetics and molecular biology, and the capability to express high levels of recombinant proteins [29, 30]. However, There are some critical disadvantages for this system, including the inability to perform post-translational modifications (such as glycosylation), improper recombinant protein folding and secretory mechanisms, as well as inefficient or low efficient disulfide bond formation. Another useful expression system is a mammalian expression system. Although protein produced in mammalian expression systems lacks the limitations of bacterial expression system, the main disadvantage of mammalian systems is their need for expensive cell culture[31]. However, baculovirus is one of the common systems for recombinant production of proteins. Here, for the first time, we successfully expressed VEGF121 in the baculovirus system. In addition, the ability of VEGF to stimulate endothelial cells was evaluated by the MTT assay.

Conflict of interest statment

The authors declare no conflict of interest.

Acknowlegments

This work was supported financially by Iran National Science Foundation (INSF), grant number 91004026.

[1] Krishna Priya S, Nagare RP, Sneha VS, Sidhanth C, Bindhya S, Manasa P, Ganesan TS. Tumour angiogenesis-Origin of blood vessels.Int J Cancer2016; 139(4): 729-35.

[2] Shimizu T, Hoshino Y, Miyazaki H, Sato E. Angiogenesis and microvasculature in the female reproductive organs: physiological and pathological implications.Curr Pharm Des2012; 18(3): 303-309.

[3] Li J, Hampton T, Morgan J, Simons M. Stretch-induced VEGF expression in the heart.J Clin Invest1997; 100(1): 18.

[4] Makrilia N, Lappa T, Xyla V, Nikolaidis I, Syrigos K. The role of angiogenesis in solid tumours: an overview.Eur J Intern Med2009; 20(7): 663-671.

[5] Seko Y, NISHIMURA H, Takahashi N, Ashida T, Nagai R. Serum Levels of Vascular Endothelial Growth Factor and Transforming Growth Factor-. BETA. 1 in Patients with Atrial Fibrillation Undergoing Defibrillation Therapy.Jpn Heart J2000; 41(1): 27-32.

[6] Fox SB, Gasparini G, Harris AL. Angiogenesis: pathological, prognostic, and growth-factor pathways and their link to trial design and anticancer drugs.Lancet Oncol2001; 2(5): 278-289.

[7] Carmeliet P ,Jain RK. Angiogenesis in cancer and other diseases.Nature2000; 407(6801): 249-257.

[8] Niethammer AG, Xiang R, Becker JC, Wodrich H, Pertl U, Karsten G, et al . Reisfeld RA. A DNA vaccine against VEGF receptor 2 prevents effective angiogenesis and inhibits tumor growth.Nat Med2002; 8(12): 1369-1375.

[9] Gordon MS, Mendelson DS, Kato G. Tumor angiogenesis and novel antiangiogenic strategies.Int J Cancer2010; 126(8): 1777-1787.

[10] Folkman J, Merler E, Abernathy C, Williams G. Isolation of a tumor factor responsible for angiogenesis.J Exp Med1971; 133(2): 275-288.

[11] Otrock ZK, Mahfouz RA, Makarem JA, Shamseddine AI. Understanding the biology of angiogenesis: review of the most important molecular mechanisms.Blood Cells Mol Dis2007; 39(2): 212-220.

[12] Tammela T, Enholm B, Alitalo K, Paavonen K. The biology of vascular endothelial growth factors.Cardiovasc Res2005; 65(3): 550-563.

[13] Pradeep C, Sunila E, Kuttan G. Expression of vascular endothelial growth factor (VEGF) and VEGF receptors in tumor angiogenesis and malignancies.Integr Cancer Ther2005; 4(4): 315-321.

[14] Cross MJ, Dixelius J, Matsumoto T, Claesson-Welsh L. VEGF-receptor signal transduction.Trends Biochem Sci2003; 28(9): 488-494.

[15] Olsson A-K, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling? In control of vascular function.Nat Rev Mol Cell Biol2006; 7(5): 359-371.

[16] Yue X, Tomanek,R.J. Effects of VEGF165 and VEGF121 on vasculogenesis and angiogenesis in cultured embryonic quail hearts.AmJ Physiol Heart Circ Physiol2001; 280: 2240-2247.

[17] Dulak J, Józkowicz A, Ratajska A, Szuba A, Cooke JP, Dembinska-Kiec Aldona. pSG5VEGF plasmids in vascular smooth muscle cells Vascular endothelial growth factor is efficiently synthesized in spite of low transfection efficiency of pSG5VEGF plasmids in vascular smooth muscle cells. Vasc Med2000; 5: 33-40.

[18] Kim S, Mohamedali KA, Cheung LH, Rosenblum MG. Overexpression of biologically active VEGF121 fusion proteins inEscherichia coli.J Biotechnol2007; 128: 638-647.

[19] Liu Y, Cheung LH, Thorpe P, Rosenblum M. Mechanistic studies of a novel, human fusion toxin composed of vascular endothelial growth factor (VEGF)121 and the serine protease granzyme B: Directed apoptotic events in vascular endothelial cells.Mol Cancer Ther2003; 2: 949-959.

[20] Kidd IM, Emery VC. The use of baculoviruses as expression vectors.Appl Biochem Biotechnol1993; 42(2-3): 137-159.

[21] Ghosh S, Parvez K, Banerjee K, Sarin SK, Hasnain SE. Baculovirus as mammalian cell expression vector for gene therapy: an emerging strategy.Mol Ther2002; 6(1): 5-10.

[22] Shao-Hua C, Hong-Liang S, Zuo-Hu L. Effect of temperature oscillation on insect cell growth and baculovirus replication.Appl Environ Microbiol1998; 64(6): 2237-2239.

[23] Jarvis DL, Kawar ZS, Hollister JR. Engineering N-glycosylation pathways in the baculovirus-insect cell system.Curr Opin Biotechnol1998; 9(5): 528-533.

[24] Kazemi-Lomedasht F, Behdani M, Pooshang Bagheri K, Habibi Anbouhi M, Abolhassani M, Khanahmad H, et al. Mirzahoseini H. Expression and purification of functional human vascular endothelial growth factor-a121; the most important angiogenesis factor.Adv Pharm Bull2014; 4(4): 323-328.

[25] Carmo A, Giovanni D, Corrêa T, Martins L, Stocco R, Suazo C, et al. Mendon?a R. Expression of an antiviral protein fromLonomia obliquahemolymph in baculovirus/insect cell system.Antiviral Res2012; 94(2): 126-130.

[26] Kazemi-Lomedasht F, Behdani M, Bagheri KP, Habibi-Anbouhi M, Abolhassani M, Arezumand R, et al . Mirzahoseini H. Inhibition of angiogenesis in human endothelial cell using VEGF specific nanobody.Mol Immunol2015; 65(1): 58-67.

[27] Assenberg R, Wan PT, Geisse S, Mayr LM. Advances in recombinant protein expression for use in pharmaceutical research.Curr Opin Struct Biol2013; 23(3): 393-402.

[28] LaVallie ER, McCoy JM. Gene fusion expression systems inEscherichia coli.Curr Opin Biotechnol1995; 6(5): 501-506.

[29] Chen R. Bacterial expression systems for recombinant protein production:E. coliand beyond.Biotechnol Adv2012; 30(5): 1102-1107.

[30] Shokrollahi N, Shahbazzadeh D, Pooshang-Bagheri K, Habibi-Anbouhi M, Jahanian-Najafabadi A, Behdani M. A model to study the phenotypic changes of insect cell transfection by copepod super green fluorescent protein (cop-GFP) in baculovirus expression system.Iran Biomed J2015.

[31] Walsh G. Biopharmaceutical benchmarks 2010.Nature Biotechnol2010; 28(9): 917-924.

ment heading

10.1016/j.apjtm.2016.09.012

Nastaran Mohseni, Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran.

?Corresponding authors: M. Behdani, Biotechnology Research Center, Venom & Biotherapeutics Molecules Lab, Pasteur Institute of Iran, Tehran, Iran.

Tel/Fax: +98 2166480780

E-mail: Behdani73042@yahoo.com, Behdani@pasteur.ac.ir

Recombinant bacmid

猜你喜歡
產品品質低端兵團
兵團記憶
兵團記憶
滾揉技術在肉制品加工中的應用研究進展
冷軋MES系統對生產的執行與支撐
如何科學合理使用植物生長調節劑
淺議中職學?!暗投恕睂W生的教育
SILICON SAGA
任正非要求華為重視低端機
服裝核心競爭力要素研究
兵團在
91香蕉高清国产线观看免费-97夜夜澡人人爽人人喊a-99久久久无码国产精品9-国产亚洲日韩欧美综合