African Journal of
Biotechnology

  • Abbreviation: Afr. J. Biotechnol.
  • Language: English
  • ISSN: 1684-5315
  • DOI: 10.5897/AJB
  • Start Year: 2002
  • Published Articles: 12486

Full Length Research Paper

Expression and purification of human IL-2 protein from Escherichia coli

Ramadan Hassan
  • Ramadan Hassan
  • Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, 60 Elgomhoria Street, 35516, Mansoura, Egypt.
  • Google Scholar
Mohammed El-Mowafy
  • Mohammed El-Mowafy
  • Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, 60 Elgomhoria Street, 35516, Mansoura, Egypt.
  • Google Scholar
Eman Samy Zaher
  • Eman Samy Zaher
  • Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, 60 Elgomhoria Street, 35516, Mansoura, Egypt.
  • Google Scholar


  •  Received: 16 February 2018
  •  Accepted: 09 April 2018
  •  Published: 25 April 2018

 ABSTRACT

Human interleukin 2 protein (IL-2) is an important cytokine found to be elevated in several types of cancer. A synthetic DNA sequence of the cDNA of mature IL-2 protein was cloned into the pRSET-B expression vector. The expressed IL-2 protein in Escherichia coli [BL21] was associated with the formation of insoluble inclusion bodies (IBs). The effect of different cultivation conditions (temperature, isopropyl-β-D-thiogalactoside concentration, and early harvest of cells) together with the incorporation of single or dual His-tag on the formation of IBs of the expressed protein was investigated. Yet, expression of soluble IL-2 was not achieved under any of the investigated conditions. A simple protocol for rapid and effective solubilization of these IBs was optimized. Using this protocol, together with subsequent purification using ion metal affinity chromatography, a purified His-IL2 protein was obtained in a yield of 5.1 mg/cell pellet of 1 L culture. In conclusion, the effect of different expression conditions on the solubility behavior of an expressed eukaryotic protein in E. coli was investigated using human IL-2 as a model protein. Moreover, the purified expressed protein could be used as a positive control in early diagnosis of tumors and in cancer research in Egypt.

Key words: Human interleukin-2, protein expression, inclusion bodies.

 


 INTRODUCTION

Interleukins are proteins that play a vital role in intercellular communication among leukocytes. Such proteins are cytokines that are classified into different classes Il-1 to Il-38 (Akdis et al., 2016), in addition to IL-39 (Wang et al., 2016). Human interleukin-2 (IL-2) is a protein produced by human lymphocytes that have been stimulated by mitogens or antigens (Ju et al., 1987). IL-2 is first synthesized as a precursor polypeptide of 153 amino acids followed by cleavage of the first 20-amino acids signal peptide resulting in the production of the mature secreted IL-2 protein (133 amino acids, 15 kDa) (Schoner et al., 1992). The mature protein is a potent cytokine that regulates innate lymphoid cells, acts as a B-cell growth factor, promotes antibody synthesis, and induces proliferation and differentiation of natural killer cells (NK) to enhance their cytolytic activities (Roediger et al., 2015). Additionally, it has been used in anti-tumor therapy (Sengupta et al., 2008; Janik et al., 1993), and intreatment of patients with disseminated cancers (Schoner et al., 1992). Several studies showed the increased expression levels of IL-2 in different types of cancer e.g. prostate cancer (Royuela et al., 2000), stomach and renal cancer (Lin et al., 1995), squamous cell carcinomas of the head and neck (Reichert et al., 1998), and neuroblastic tumors (Ridings et al., 1995). Therefore, the elevated expression levels of such cytokine could be used as a diagnostic marker for detection of actively proliferating tumors (Garcia-Tunon et al., 2004). Recombinant protein expression in Escherichia coli is useful for different purposes. (i) The production of vaccines such as the expression of chicken anemia virus capsid protein VP1 (Lee et al., 2011). (ii) The production of protein that could be used in serodiagnosis of infections e.g. poliovirus (Uma et al., 2016), and Brucella melitensis (Cloeckaert et al., 2001). (iii) Production of purified proteins that are sold by research companies to be used as positive control in different research experiments e.g. recombinant IL-2 in Western blot and ELISA (Gehman and Robb, 1984). The use of E. coli as an expression system has many advantages e.g. rapid transformation process, high yield of expression, and the whole expression is less expensive in comparison with other hosts (Rosano and Ceccarelli, 2014). However, the higher expression rates of recombinant proteins in E. coli are often accompanied with the formation of insoluble aggregates of the target protein called inclusion bodies (IBs) (Singh et al., 2015). The aim of this study was to express the recombinant IL-2 protein in considerable amounts that could be used as a positive control for early diagnosis of tumors and in cancer research in Egypt. Another aim was to investigate the effect of different expression conditions on the solubility behavior of eukaryotic protein expression in E. coli using human IL-2 as a model protein.


 MATERIALS AND METHODS

Bacterial strains and construction of recombinant plasmids harboring IL-2 gene
 
The E. coli strain Top10 (Thermo Fisher Scientific) was used for cloning, propagation and maintenance of the constructed plasmids; while the E. coli strain BL21 (DE3) [Invitrogen] was used for recombinant protein expression. Both strains were transformed using CaCl2-heat shock method (El-Mowafy et al., 2013). Selection of positive transformants was performed on Luria-Bertani (LB) medium agar plates containing ampicillin (100 µg/ml). Positive clones were further cultivated in LB medium containing ampicillin (100 µg/ml). Glycerol stocks of all strains were prepared and stored at -80°C till further use (El-Mowafy et al., 2013). The cDNA of human IL-2 precursor (462 bases, 153 amino acids) (Devos et al., 1983) was synthesized and provided by Macrogen Inc (Korea) in a bacterial cloning plasmid (pl-IL2) with ampicillin as a selection marker. The primers IL2-F1and IL2-R1 (Table 1) were used for cloning of mature IL-2 (402 bp, 133 amino acids) from pl-IL2 into pRSET-B expression vector (Invitrogen). The amplified IL-2 gene and pRSET-B vector were digested with BamHI (NEB, UK) and HindIII (NEB, UK) followed by ligation with T4 DNA ligase enzyme (NEB) to form the plasmid pl-His-IL2. The cloned IL-2 gene was in frame with the nucleotides encoding the N-terminal 6 His-tag, which is already introduced by Invitrogen in the pRSET-B vector, resulting in the expression of His-IL2 protein. 
 
 
Similarly, another version of IL-2 was also cloned into pRSET-B vector as a BamHI/HindIII fragment from pl-IL2 using the primers IL2-F1 and IL2-R2 (Table 1) into pRSET-B expression vector to obtain the plasmid pl-His-IL2-His. The expressed Il-2 protein (His-IL2-His) from the latter plasmid was dual His-tagged at both terminals. This was achieved by introducing the nucleotides encoding the C-terminal 6 His-tag at the primer IL2-R2, in addition to the cloning of the BamHI/HindIII-digested polymerase chain reaction (PCR) fragment of IL-2 in frame with the nucleotides encoding the N-terminal 6 His-tag of the pRSET-B vector. Positive clones were confirmed by restriction digestion with BamHI and HindIII. All the cloning steps were performed using Phusion High-Fidelity DNA Polymerase (NEB, UK). All PCR reactions were performed in MultiGene™ Mini Personal Thermal Cycler (Labnet, Multigene gradient, Foster City, CA). The PCR products were electrophoresed on a 1.5% agarose gel and visualized by staining with ethidium bromide. The size of DNA fragments was confirmed using Hyperladder™ 100 bp (Bioline, England) and 1 kb DNA ladder (NEB, UK).
 
Expression of IL-2 in E. coli BL21 at different conditions
 
LB medium containing ampicillin (100 µg/ml) was inoculated from the glycerol stock of E. coli BL21 transformed with pRSET-B, pl-His-IL2 or pl-His-IL2-His plasmids and incubated at 37°C with shaking (200 rpm). After overnight cultivation, subculturing into flasks containing 50 ml fresh medium supplemented with the antibiotic was performed so that the starting OD600nm was 0.5 (Ma et al., 2006). The expression was induced with 0.5 mM isopropyl-β-D-thiogalactopyranoside (IPTG). Preliminary time course screening of expression was performed by harvesting 1.5 ml culture after 3, 4, and 5 h of induction followed by centrifugation at 878 × g for 5 min. The cell pellet was suspended in 30 µl 5x SDS gel loading buffer (0.25 M Tris-HCl, pH=6.8, 40% Glycerol, 8% SDS, 2.9 mM β-mercaptoethanol and 0.1% bromophenol blue) followed by heating at 95°C for 10 min (Laemmli UK, 1970). The resulting denaturated protein samples were analyzed by 15% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), before staining with Coomassie Brilliant Blue (Laemmli UK, 1970). The size of protein bands was confirmed using chromatein prestained protein marker (Biosepes, China) or Roti®-Mark standard protein marker (Roth, Germany). For efficient lysis of the cell pellet after IPTG induction, sonication followed by enzymatic lysis was performed. Briefly, the whole cell pellet after IPTG induction was suspended in 3x volume lysis buffer (10 mM sodium phosphate buffer pH 7, 5 mM NaCl, 5 mM KCl, and protease inhibitor [Complete Ultra, mini, EDTA free, Roche]) (El-Mowafy et al., 2013). The bacterial suspension was subjected to sonication using a Soniprep (150 Sonicator TM-182; UK) in intervals of 30 s sonication and 30 s without sonication for a total of 5 min, while immersed in ice. To allow for enzymatic lysis, lysozyme solution (2 mg/ml) in 20 mM Tris-HCl (pH 8) was added to the sonicated cell suspension in a ratio of 3x volume of the initial cell pellet followed by immersion in ice for 30 min. Benzonase enzyme (EMD Millipore, USA) was added to the mixture to decrease the viscosity of the lysate by degradation of nucleic acids (Final concentration 25 U/ml). The soluble cell lysate (soluble protein extract) was obtained by centrifugation of the whole cell lysate at 11200×g for 10 min, at 4°C, from which an aliquot was denaturated by heating in 5x SDS gel loading. Another aliquot was taken for the total protein extract (soluble and insoluble protein extract) before centrifugation, as a control, and was denaturated by heating in 5x SDS gel loading (Laemmli UK, 1970). The expression of the recombinant IL-2 protein in the total protein extract and the soluble cell lysate was investigated at different cultivation conditions by SDS-PAGE Coomassie staining. Such conditions included induction of expression at lower concentration of IPTG (0.25 mM), harvesting at earlier time of expression (3 h after IPTG induction), and expression of the protein at lower incubation temperature (35 or 30°C) (Sorensen and Mortensen, 2005a).
 
Recovery, purification and solubilization of IBs to obtain soluble purified IL-2 protein
 
The cell pellet from 100 ml culture was collected after IPTG induction and subjected to lysis by sonication and lysozyme enzyme before separation of the soluble protein extract as mentioned earlier. The protocol was carried out as described previously (Schoner et al., 1992) with few modifications. Briefly, the pellet (containing the disrupted cells, insoluble cellular proteins and IL-2 inclusions bodies) was suspended in 100 mM Tris-HCl (pH 8.5) containing triton 1% and urea 1 M. The mixture was left on ice for 10 min followed by centrifugation at 11200 ×g for 10 min at 4°C. Washing of the pellet with triton and urea was repeated for additional 2 times, before washing with 100 mM Tris-HCl, pH 8.5. For solubilization of IBs, the pellet was resuspended in equal volume of 8 M guanidine HCl in 100 mM Tris-HCl (pH 8.5) and left in ice for 1 h with gentle shaking. The mixture was centrifuged at 11200 × g for 10 min at 4°C. The supernatant containing the solubilized IBs was subjected to Protino™ Ni-NTA agarose beads (Macherey-Nagel, Germany) to fish the His-tagged-IL2 protein according to the manufacturer’s instructions. After elution of the Ni+2 binded His-tagged IL2 protein with imidazole (500 mM), excess imidazole concentration was removed by ultrafiltration with 100 mM Tris-HCl, pH 7 using Roti®-Spin MINI-3 centrifugal device (Roth, Germany). The purified IL-2 protein was stored at -80°C till further use (El-Mowafy et al., 2013).
 
Western blot
 
Western blot analysis of the expressed IL-2 proteins was performed using diluted (1:1000) 6-Histidine Epitope Tag Antibody [HRP] (Novus Biologicals, USA) as previously mentioned in El-Mowafy et al. (2013), except that the His-tagged protein bands were visualized by incubation of the membrane with tetramethylbenzidine (TMB) substrate solution (Sigma Aldrich, USA) at room temperature for few minutes before capturing of photos (Shaaban et al., 2015).
 
Determination of protein content
 
 
The concentration of the purified IL-2 protein was measured using Bradford protein assay kit (Biospes, China) according to the manufacturer’s instructions. The protein concentration was calculated from the standard curve of bovine serum albumin (positive control) after measurement of the absorbance at 595 nm using ELx808™ Absorbance Microplate Reader (Biotek Instruments Inc.,Winooski, VT) (Shaaban et al., 2015). The buffer containing the purified IL-2 protein (100 mM Tris-HCl, pH 7) was used as blank (negative control).

 


 RESULTS

PCR and cloning
 
The DNA sequence of mature IL-2 (402 bp) was successfully amplified from pl-IL2 plasmid (Figure 1A) and cloned into the pRSET-B vector, to allow for the bacterial expression of either the N-terminal His-tagged IL-2 (His-IL-2 protein from pl-His-IL2 plasmid) or the dually His-tagged IL-2 (His-IL2-His protein from pl-His-IL2-His plasmid). Cloning of both versions of IL-2 was confirmed by restriction digestion of the constructed plasmids (Figure 1B).
 
 
 
 
Recombinant expression of His-IL2 and His-IL2-His purified proteins
 
 
Bacterial expression of both versions of IL-2 protein was successfully detected at different time points following IPTG induction (Figure 2). Maximum production of the recombinant protein was observed after 4 h of induction (Figure 2). Therefore, further expression experiments were performed after 4 h of induction for both types of expressed IL-2 protein, unless otherwise specified. Formation of IBs was detected in both versions of the expressed IL-2 by SDS-PAGE of the total protein extract and the soluble lysate of the transformed E. coli BL21 cells. The presence of the target protein band in the total protein extract, but not in the soluble lysate, confirmed the formation of insoluble IBs (lanes T and S in Figure 3A and B). Purification and solubilization of IBs of His-IL2 and His-IL2-His proteins was successfully performed as indicated by SDS-PAGE Coomassie staining (Figure 3A and B) and Western blot detection using anti-histidine tag monoclonal antibody (Figure 3C).

 

 
Investigation of the effect of different expression conditions on the formation of insoluble IBs
 
Different expression conditions were used to avoid the formation of insoluble IBs. Decreasing the concentration  of IPTG to 0.25 mM (Figure 4A), earlier harvest of the induced cells (3 h of induction, Figure 4B), and decreasing the incubation temperature to 35 (Figure 4C) or 30°C (Figure 4D) did not help to avoid the formation of the IBs. This was indicated by the presence of the expressed His-IL-2 in the total protein extract, but not in the soluble lysate (lanes T and S in Figure 4).
 
 
 
 
Quantification of the yield of the purified recombinant IL-2 proteins
 
 
After purification of both versions of IL-2 protein via immobilized metal affinity chromatography of the solubilized inclusion bodies, the concentrations of purified proteins were determined. The yield of His-IL2 and His-IL-2-His purified proteins was 5.1 and 4.5 mg/cell pellet of 1 L culture, respectively.

 


 DISCUSSION

Early studies on human IL-2 protein were restricted to the protein purified from human T-cell leukemia cell line (Jurkat) (Robb et al., 1983; Stern et al., 1984). After recombinant protein technology, IL-2 was expressed after isolation of IL-2 mRNA from human leukemic T-cell line, followed by cloning of the complementary cDNA into a suitable vector. The cloned IL-2 was expressed in different hosts such as E. coli (Ju et al., 1987; Devos et al., 1983; Rosenberg et al., 1984), insect cells (Smith et al., 1985), and monkey COS cells (Taniguchi et al., 1983). Most of the expressed IL-2 in E. coli was reported to be insoluble (forms IBs), however, the recombinant IL-2 was still retaining its biological activity even after extraction from SDS polyacrylamide gel (Devos et al., 1983). In this study, the DNA sequence of the cloned IL-2 gene was derived from a synthesized DNA sequence rather than extraction of mRNA from human leukemic T-cell line and subsequent conversion into cDNA. An N-terminal His-tagged IL-2 (His-IL-2) protein was expressed in E. coli BL21 cells. Insoluble IBs of the recombinant His-IL-2 protein was formed, which was indicated by the absence of the target protein in the soluble lysate of the cells, despite its presence in the total protein extract (Figure 3A). A simple protocol was optimized for the solubilization of the insoluble aggregates of both versions of the expressed IL-2 protein (Figure 3). Such protocol depends on washing of the IBs with low concentrations of tritone (1%) and urea (1 M) to get rid of cell wall and cell membrane components without solubilization of the IBs. The detergent guanidine hydrochloride (8 M) was used for solubilization of the IBs, followed by fishing of the His-tagged IL2 protein using Ni+2 agarose beads to obtain the purified recombinant proteins (Figure 3) (Palmer and Wingfield 2004).
 
Different strategies were followed to prevent formation of IBs of the expressed recombinant His-IL2 protein. Incorporation of fusion tags was reported to increase the solubility of the expressed protein and subsequently could prevent formation of IBs (Costa et al., 2014; Sorensen and Mortensen, 2005b). Therefore, a dual His-tagged IL-2 protein (His-IL-2-His) was expressed to decrease the chance of IBs formation. However, the expression of this version of IL-2 protein (His-IL-2-His) was also associated with the formation of IBs (Figure 3B).  High expression rates of recombinant proteins in E. coli contribute greatly in IBs formation as described previously by Gatti-Lafranconi et al. (2011). Additionally, the use of high incubation temperature and high IPTG concentration often leads to a high expression rate and subsequent formation of IBs (Singh et al., 2015). Therefore, attempt was made to prevent the formation of IBs by decreasing the cultivation temperature to 35 or 30°C, and by decreasing the concentration of the expression-inducing agent to 0.25 mM. Additionally, the cells were harvested after 3 h of IPTG induction before they enter in the stage of high rate of protein expression. Nevertheless, none of these trials were useful to avoid the formation of IBs (Figure 4).


 CONCLUSION

The impact of the different expression conditions of IL-2 protein on the formation of IBs was determined. Yet, IBs were still forming under all the investigated conditions. This study provides an inexpensive protocol for the production of purified human IL-2 protein. The purified human IL-2 protein produced by this work could be used as a positive control for early diagnosis of tumors and in cancer research in Egypt. Additionally, the biological activity of the purified protein, after trying different refolding conditions, will be investigated in a future project. 


 CONFLICT OF INTERESTS

The authors have not declared any conflict of interests.

 



 REFERENCES

Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, Duan S, Eiwegger T, Eljaszewicz A, Ferstl R, Frei R (2016). Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 138(4):984-1010.
Crossref

 

Cloeckaert A, Baucheron S, Vizcaino N, Zygmunt MS (2001). Use of recombinant BP26 protein in serological diagnosis of Brucella melitensis infection in sheep. Clin. Diagn. Lab. Immunol. 8(4):772-775.
Crossref

 

Costa S, Almeida A, Castro A, Domingues L (2014). Fusion tags for protein solubility, purification and immunogenicity in Escherichia coli: the novel Fh8 system. Front. Microbiol. 5:63.
Crossref

 

Devos R, Plaetinck G, Cheroutre H, Simons G, Degrave W, Tavernier J, Remaut E, Fiers W (1983). Molecular cloning of human interleukin 2 cDNA and its expression in E. coli. Nucleic Acids Res. 11(13):4307-4323.
Crossref

 

El-Mowafy M, Bahgat MM, Bilitewski U (2013). Deletion of the HAMP domains from the histidine kinase CaNik1p of Candida albicans or treatment with fungicides activates the MAP kinase Hog1p in S. cerevisiae transformants. BMC Microbiol. 13:209.
Crossref

 

Garcia-Tunon I, Ricote M, Ruiz A, Fraile B, Paniagua R, Royuela M (2004). Interleukin-2 and its receptor complex (alpha, beta and gamma chains) in in situ and infiltrative human breast cancer: an immunohistochemical comparative study. Breast Cancer Res. 6(1):R1-7.
Crossref

 

Gatti-Lafranconi P, Natalello A, Ami D, Doglia SM, Lotti M (2011). Concepts and tools to exploit the potential of bacterial inclusion bodies in protein science and biotechnology. FEBS J. 278(14):2408-2418.
Crossref

 

Gehman LO, Robb RJ (1984). An ELISA-based assay for quantitation of human interleukin 2. J. Immunol. Methods 74(1):39-47.
Crossref

 

Janik JE, Sharfman WH, Smitht JW, Longo DL, Sznol M, Urba WJ, Figlin R, Pierce WC, Bukowski RM, Fyfe G, Belldegrun A (1993). Erythropoietin production. A potential marker for interleukin-2/interferon-responsive tumors. Cancer 72(9):2656-2659.
Crossref

 

Ju G, Collins L, Kaffka KL, Tsien WH, Chizzonite R, Crowl R, Bhatt R, Kilian PL (1987). Structure-function analysis of human interleukin-2. Identification of amino acid residues required for biological activity. J. Biol. Chem. 262(12):5723-5731.

 

Laemmli UK (1970). Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227(5259):680-685.
Crossref

 

Lee MS, Hseu YC, Lai GH, Chang WT, Chen HJ, Huang CH, Lee MS, Wang MY, Kao JY, You BJ, Lin WH (2011). High yield expression in a recombinant E. coli of a codon optimized chicken anemia virus capsid protein VP1 useful for vaccine development. Microb. Cell Fact. 10:56.
Crossref

 

Lin WC, Yasumura S, Suminami Y, Sung MW, Nagashima S, Stanson J, Whiteside TL (1995). Constitutive production of IL-2 by human carcinoma cells, expression of IL-2 receptor, and tumor cell growth. J. Immunol. 155(10):4805-4816.

 

Ma X, Zheng W, Wei D, Ma Y, Wang T, Wang J, Liu Q, Yang S (2006). Construction, expression, and purification of HIV-TAT-survivin (T34A) mutant: a pro-apoptosis protein in Escherichia coli. Protein Expr. Purif. 47(1):36-44.
Crossref

 

Palmer I, Wingfield PT (2004). Preparation and extraction of insoluble (inclusion-body) proteins from Escherichia coli. Current protocols in protein science, Chapter 6: Unit 63.

 

Reichert TE, Watkins S, Stanson J, Johnson JT, Whiteside TL (1998). Endogenous IL-2 in cancer cells: a marker of cellular proliferation. J. Histochem. Cytochem. 46(5):603-611.
Crossref

 

Ridings J, Macardle PJ, Byard RW, Skinner J, Zola H (1995). Cytokine receptor expression by solid tumours. Ther. Immunol. 2(2):67-76.

 

Robb RJ, Kutny RM, Chowdhry V (1983). Purification and partial sequence analysis of human T-cell growth factor. Proc. Natl. Acad. Sci. USA, 80(19):5990-5994.
Crossref

 

Roediger B, Kyle R, Tay SS, Mitchell AJ, Bolton HA, Guy TV, Tan SY, Forbes-Blom E, Tong PL, Köller Y, Shklovskaya E (2015). IL-2 is a critical regulator of group 2 innate lymphoid cell function during pulmonary inflammation. J. Allergy Clin. Immunol. 136(6):1653-1663.
Crossref

 

Rosano GL, Ceccarelli EA (2014). Recombinant protein expression in Escherichia coli: advances and challenges. Front. Microbiol. 5:172.
Crossref

 

Rosenberg SA, Grimm EA, Mcgrogan M, Doyle M, Kawasaki E, Koths K, Mark DF (1984). Biological activity of recombinant human interleukin-2 produced in Escherichia coli. Science 223(4643):1412-1414.
Crossref

 

Royuela M, De Miguel MP, Bethencourt FR, Fraile B, Arenas MI, Paniagua R (2000). IL-2, its receptors, and bcl-2 and bax genes in normal, hyperplastic and carcinomatous human prostates: immunohistochemical comparative analysis. Growth Factors 18(2):135-146.
Crossref

 

Schoner RG, Ellis LF, Schoner BE (1992). Isolation and purification of protein granules from Escherichia coli cells overproducing bovine growth hormone. 1985. Biotechnology (Reading, Mass.) 24:349-352.

 

Sengupta P, Meena K, Mukherjee R, Jain SK, Maithal K (2008). Optimized conditions for high-level expression and purification of recombinant human interleukin-2 in E. coli. Indian J. Biochem. Biophys. 45(2):91-97.

 

Shaaban MI, Abdelmegeed E, Ali YM (2015). Cloning, Expression, and Purification of Recombinant Uricase Enzyme from Pseudomonas aeruginosa Ps43 Using Escherichia coli. J. Microbiol. Biotechnol. 25(6):887-892.
Crossref

 

Singh A, Upadhyay V, Upadhyay AK, Singh SM, Panda AK (2015). Protein recovery from inclusion bodies of Escherichia coli using mild solubilization process. Microb. Cell Fact. 14:41.
Crossref

 

Smith GE, Ju G, Ericson BL, Moschera J, Lahm HW, Chizzonite R, Summers MD (1985). Modification and secretion of human interleukin 2 produced in insect cells by a baculovirus expression vector. Proc. Natl. Acad. Sci. USA. 82(24):8404-8408.
Crossref

 

Sorensen HP, Mortensen KK (2005a). Soluble expression of recombinant proteins in the cytoplasm of Escherichia coli. Microb. Cell Fact. 4(1):1.
Crossref

 

Sorensen HP, Mortensen KK (2005b). Advanced genetic strategies for recombinant protein expression in Escherichia coli. J. Biotechnol. 115(2):113-128.
Crossref

 

Stern AS, Pan YC, Urdal DL, Mochizuki DY, Dechiara S, Blacher R, Wideman J, Gillis S (1984). Purification to homogeneity and partial characterization of interleukin 2 from a human T-cell leukemia. Proc. Natl. Acad. Sci. USA. 81(3):871-875.
Crossref

 

Taniguchi T, Matsui H, Fujita T, Takaoka C, Kashima N, Yoshimoto R, Hamuro J (1983). Structure and expression of a cloned cDNA for human interleukin-2. Nature 302(5906):305-310.
Crossref

 

Uma M, Rao PP, Nagalekshmi K, Hegde NR (2016). Expression and purification of polioviral proteins in E. coli, and production of antisera as reagents for immunological assays. Protein Expr. Purif. 128:115-122.
Crossref

 

Wang X, Liu X, Zhang Y, Wang Z, Zhu G, Han G, Chen G, Hou C, Wang T, Ma N, Shen B (2016). Interleukin (IL)-39 [IL-23p19/Epstein-Barr virus-induced 3 (Ebi3)] induces differentiation/expansion of neutrophils in lupus-prone mice. Clin. Exp. Immunol. 186(2):144-156.
Crossref

 




          */?>