• Title/Summary/Keyword: MBP fusion protein

Search Result 24, Processing Time 0.025 seconds

Single-Step Purification of Proteins of Interest from Proteolytically Cleaved Recombinant Maltose-binding Protein (MBP) Fusion Proteins by Selective Immunoprecipitation of MBP

  • Park, Jung-Hyun;Na, Shin-Young;Lee, Dong-Gun;Han, Byoung-Don;Kim, Kil-Lyong
    • Biotechnology and Bioprocess Engineering:BBE
    • /
    • v.3 no.2
    • /
    • pp.82-86
    • /
    • 1998
  • The maltose binding protein (MBP) fusion protein system is a versatile tool to express and isolate recombinant proteins in E. coli. In this system, MBP fusion proteins are efficiently isolated from whole cell lysate using amylose conjugated agarose beads and then eluted by competition with free maltose. Since MBP is a rather large molecule (∼42 kDa), for further experiments, the MBP part is usually proteolytically cleaved from the fusion protein and subsequently removed by ion-exchange chromatography or rebinding to amylose columns after washing out excess and MBP-bound maltose. In the present study, we have developed an improved method for the removal of cleaved MBP, which is advantageous over conventional methods. In this method, factor Xa cleaved MBP fusion proteins were incubated with Sepharose beads conjugated with MBP specific monoclonal antibodies and then precipitated buy centrifugation, resulting in highly purified proteins in the supernatant.

  • PDF

Soluble Expression of Human Angiostatin and Endostatin by Maltose Binding Protein (MBP) Fusion in E. coli (Maltose Binding Protein 융합단백질에 의한 인간유래의 앤지오스타틴과 앤도스타틴의 대장균에서 수용성 단백질발현)

  • Paek, Seon-Yeol;Choi, Shin-Geon
    • Journal of Industrial Technology
    • /
    • v.28 no.B
    • /
    • pp.59-63
    • /
    • 2008
  • Rapid production of therapeutic proteins such as angiostatin and endostatin angiogenic inhibititors has been highly demanded for cancer treatment. In this regard, recombinant human angiostatin and endostatin were successfully expressed as soluble forms by maltose binding protein (MBP)-mediated fusion expression in Escherichia coli. PCR amplified, angiostatin and endostatin genes from human placenta cDNA library were inserted into an expression vector pMAL-c2e to construct prokaryotic expression vectors, pMAL-c2e/AS and pMAL-c2e/ES, respectively. Recombinant angiostatin and endostatin were efficiently expressed in E. coli origami (DE3) after IPTG induction and protein expression were confirmed by SDS-PAGE analyses. The expressed recombinant proteins were purified near homogenity using an amylose affinty column chromatography. In contrast that previous E. coli expressions were all insoluble, our results first time demonstrated that MBP fused human angiostatin and endostatin were soluble in E. coli.

  • PDF

Expession of the Recombinant Klebsiella aerognes UreF Protein as a MalE Fusion

  • Kim, Keun-Young;Yang, Chae-Ha;Lee, Mann-Hyung
    • Archives of Pharmacal Research
    • /
    • v.22 no.3
    • /
    • pp.274-278
    • /
    • 1999
  • Expression of the active urease of the enterobacterium, Klebsiella aerogens, requires the presence of the accessory genes (ureD, ureE, ureF, and ureG) in addition to the three structural genes (ureA, ureB, and ureC). These accessory genes are involved in functional assembly of the nickel-metallocenter for the enzyme. Characterization of ureF gene has been hindered, however, since the UreF protein is produced in only minute amount compared to other urease gene products. In order to overexpress the ureF gene, a recombinant pMAL-UreF plasmid was constructed from which the UreF was produced as a fusion with maltose-binding protein. The MBP-UreF fusion protein was purified by using an amylose-affinity column chromatography followed by an anion exchange column chromatography. Polyclonal antibodies raised against the fusion protein were purified and shown to specifically recognize both MBP and UreF peptides. The UreF protein was shown to be unstable when separated from MBP by digestion with factor Xa.

  • PDF

The Soluble Expression of the Human Renin Binding Protein Using Fusion Partners: A Comparison of ubquitin, Thioredoxin, Maltose Binding Protein-and NusA

  • Lee, Chung;Lee, Sun-Gu;Saori Takahashi;Kim, Byung-Gee
    • Biotechnology and Bioprocess Engineering:BBE
    • /
    • v.8 no.2
    • /
    • pp.89-93
    • /
    • 2003
  • human renin binding protein (hRnBp), showing N-acetylglucosamine-2-epimerase activity, was over-expressed in E. coli, but was mainly present as an inclusion body. To improve its solubility and activity, ubiquitin (Ub), thioredoxin (Trx), maltose binding protein (MBP) and NusA, were used as fusion partners. The comparative solubilities of the fusion proteins were, from most to least soluble: NusA, MBP, Trx, Ub. Only the MBP fusion did not significantly reduce the activity of hRnBp, but enhanced the stability. The Origami (DE3), permitting a more oxidative environment for the cytoplasm in E. coli; helped to increase its functional activity.

Soluble Expression and Purification of Receptor Activator of Nuclear Factor-Kappa B Ligand Using Escherichia coli

  • Park, Sol-Ji;Lee, Se-Hoon;Kim, Kwang-Jin;Kim, Sung-Gun;Kim, Hangun;Choe, Han;Lee, Sang Yeol;Yun, Jung-Mi;Cho, Jae Youl;Chun, Jiyeon;Choi, Kap Seong;Son, Young-Jin
    • Journal of Microbiology and Biotechnology
    • /
    • v.25 no.2
    • /
    • pp.274-279
    • /
    • 2015
  • Receptor activator of nuclear factor-kappa B ligand (RANKL) is a critical factor in osteoclastogenesis. It makes osteoclasts differentiate and multinucleate in bone remodeling. In the present study, RANKL was expressed as a soluble maltose binding protein (MBP)-fusion protein using the Escherichia coli maltose binding domain tag system (pMAL) expression vector system. The host cell E. coli DH5α was cultured and induced by isopropyl β-D-1-thiogalactopyranoside for rRANKL expression. Cells were disrupted by sonication to collect soluble MBP-fused rRANKL. The MBP-fusion rRANKL was purified with MBP Trap affinity chromatography and treated with Tobacco Etch Virus nuclear inclusion endopeptidase (TEV protease) to remove the MBP fusion protein. Dialysis was then carried out to remove binding maltose from the cleaved rRANKL solution. The cleaved rRANKL was purified with a second MBP Trap affinity chromatography to separate unsevered MBP-fusion rRANKL and cleaved MBP fusion protein. The purified rRANKL was shown to have biological activity by performing in vitro cell tests. In conclusion, biologically active rRANKL was successfully purified by a simple two-step chromatography purification process with one column.

Expression of an Antimicrobial Peptide Magainin by a Promoter Inversion System

  • Lee, Jae-Hyun;Hong, Seung-Suh;Kim, Sun-Chang
    • Journal of Microbiology and Biotechnology
    • /
    • v.8 no.1
    • /
    • pp.34-41
    • /
    • 1998
  • A method was developed for the controlled expression of an antimicrobial peptide magainin in Escherichia coli. A series of concatemeric magainin genes was constructed with a gene amplification vector, and fused to the 3'end of malE gene encoding the affinity ligand, E. coli maltose-binding protein (MBP). The construct directed the synthesis of the fusion protein with the magainin polypeptide fused to the C-terminus of MBP. The fusion protein was expressed in a tightly regulatable expression system which was under the control of an invertible promoter. The MBP-fused magainin monomer was expressed efficiently. However, the expression level of the MBP-fused magainin in E. coli decreased with the increasing size of multimers possibly because of the transcription and translation inhibition by the multimeric peptides. After purification using an amylose affinity column, the fusion protein was digested by factor Xa at a specific cleavage site between the monomers. The recombinant magainin had an antimicrobial activity identical to that of synthetic magainin. This experiment shows that a biologically active, antimicrobial peptide magainin can be produced by fusing to MBP, along with a promoter inversion vector system.

  • PDF

Comparisons of Recombinant Protein Expression in Diverse Natural Isolates of Escherichia coli

  • Jung, Yuna;Lim, Dongbin
    • Molecules and Cells
    • /
    • v.25 no.3
    • /
    • pp.446-451
    • /
    • 2008
  • We assessed heterologous protein expression in 64 strains obtained from the Escherichia coli Reference (ECOR) collection, a collection representing diverse natural E. coli populations. A plasmid generating a glutathione S-transferase and plant carbonic anhydrase fusion protein (GST-CA) under the control of the tac promoter was introduced into the ECOR strains, and the quantity of the fusion protein was determined by SDS-PAGE. The foreign protein was generated at various levels, from very high (40 strains, high producers) to very low (six strains, low producers). Immunoblotting showed that the high producers expressed approximately 250-500 times more GST-CA protein than the low producers. The results of semi-quantitative RT-PCR showed that the low producers generated mRNA levels comparable to those of the high producers, thereby suggesting that, at least in this case, inefficient translation is a major cause of the low production. We introduced a different plasmid, which expressed a maltose binding protein and plant guanylate kinase fusion protein (MBP-GK) into the six low producers. Interestingly, five of these expressed MBP-GK at very high levels. Thus, we conclude that the production of a particular protein from an expression vector can vary considerably, depending on the host strain. Strains in the ECOR collection could function as useful alternative hosts when a desired level of protein expression is not obtained from commonly used strains, such as E. coli K12 or B derivatives.

Characterization and Evaluation of a Distinct Fusion Ability in the functionally Related Cyclic Amidohydrolase Family Enzymes

  • Kim, Hak-Sung;Lee, Dong-Eun;Kim, Geun-Joong
    • Biotechnology and Bioprocess Engineering:BBE
    • /
    • v.7 no.3
    • /
    • pp.155-162
    • /
    • 2002
  • The cyclic amidohydrolase family enzymes, which include allantoinase, dihydroorotase, dihydropyrimidinase and (phenyl)hydantoinase, are metal-dependent hydrolases and play a crucial role in the metabolism of purine and pyrimidine in vivo. Each enzyme has been independently characterized, and thus well documented, but studies on the higher structural traits shared by members of this enzyme family are rare due to the lack of comparative study. Here, we report upon the expression in E. coli cells of maltose-binding protein (MBP)- and glutathione S-transferase (GST)-fused cyclic amidohydrolase family enzymes, facilitating also for both simple purification and high-level expression. Interestingly, the native quaternary structure of each enzyme was maintained even when fused with MBP and GST. We also found that in fusion proteins the favorable biochemical properties of family enzymes such as, their optimal pHs, specific activities and kinetic properties were conserved compared to the native enzymes. In addition, MBP-fused enzymes showed remarkable folding ability in-vitro. Our findings, therefore, suggest that a previously unrecognized trait of this family, namely the ability to functional fusion with some other protein but yet to retain innate properties, is conserved. We described here the structural and evolutionary implications of the properties in this family enzyme.

Caspase-3-facilitated Stoichiometric Cleavage of a Large Recombinant Polyprotein (카스파제-3 효소를 이용한 폴리-단백질의 정량적 프로세싱 분석)

  • Kim, Moonil
    • Journal of Life Science
    • /
    • v.25 no.4
    • /
    • pp.385-389
    • /
    • 2015
  • In this study, it is reported that a large polyprotein can be stoichiometrically cleaved by the use of caspase-3-dependent proteolysis. Previously, it has been shown that the proteolytic IETD motif was partially processed when treated with caspase-3, while the DEVD motif was completely cleaved. The cleavage efficiency of the DEVD-based substrate was approximately 2.0 times higher than that of the IETD substrate, in response to caspase-3. Based on this, 3 protein genes of interest were genetically linked to each other by adding two proteolytic cleavage sequences, DEVD and IETD, for caspase-3. Particularly, glutathione-S transferase (GST), maltose binding protein (MBP), and red fluorescent protein (RFP) were chosen as model proteins due to the variation in their size. The expressed polyprotein was purified by immobilized metal ion affinity chromatography (IMAC) via a hexa-histidine tag at the C-terminal end, showing 93 kDa of a chimeric GST:MBP:RFP fusion protein. In response to caspase-3, cleavage products, such as MBP:RFP (68 kDa), MBP (42 kDa), RFP (26 kDa), and GST (25 kDa), were separated from a large precursor GST:MBP:RFP (93 kDa) via SDS-PAGE. The results obtained from this study indicate that a multi-protein can be stoichiometrically produced from a large poly-protein by using proteolytic recognition motifs, such as DEVD and IETD tetra-peptides, for caspase-3.

Production of O-GlcNAc Modified Recombinant Proteins in Escherichia coli

  • LIM, KI HONG;CHANG HOON HA;HYO IHL CHANG
    • Journal of Microbiology and Biotechnology
    • /
    • v.12 no.2
    • /
    • pp.306-311
    • /
    • 2002
  • O-linked N-acetylglucosamine (O-GlcNAc) is an abundant posttranslationally modified compound in eukaryotic cells. Human O-GlcNAc transferase (OGT) was produced as a maltose binding protein (MBP) fusion protein, which showed significant catalytic activity to modify recombinant Sp1, transcription factor. To facilitate the production of O-GlcNAc modified proteins, instead of using the tedious in vitro glycosylation reaction or expression in eukaryotic cells, a MBP-fusion OGT expression vector (pACYC184-MBPOGT) was constructed using pACYC184 plasmid, which could coexist with general prokaryotic expression vectors containing ColE1 origin. By cotransforming pACYC184-MBPOGT and pGEX-2T vectors into Escherichia coli BL21, intracellular O- GlcNAcylated proteins could be obtained by a simple purification procedure. It is expected that this may be a useful tool for production of O-GlcNAc modified proteins.