KR20090025481A - Preparation method of recombinant protein by use of rpos as a fusion expression partner - Google Patents

Preparation method of recombinant protein by use of rpos as a fusion expression partner Download PDF

Info

Publication number
KR20090025481A
KR20090025481A KR1020070090368A KR20070090368A KR20090025481A KR 20090025481 A KR20090025481 A KR 20090025481A KR 1020070090368 A KR1020070090368 A KR 1020070090368A KR 20070090368 A KR20070090368 A KR 20070090368A KR 20090025481 A KR20090025481 A KR 20090025481A
Authority
KR
South Korea
Prior art keywords
protein
rpos
gene
expression vector
expression
Prior art date
Application number
KR1020070090368A
Other languages
Korean (ko)
Other versions
KR100890186B1 (en
Inventor
이지원
박진승
한경연
송종암
서혁성
안금영
이종환
이은정
권수정
Original Assignee
고려대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 고려대학교 산학협력단 filed Critical 고려대학교 산학협력단
Priority to KR1020070090368A priority Critical patent/KR100890186B1/en
Priority to PCT/KR2008/005256 priority patent/WO2009031852A2/en
Priority to US12/676,982 priority patent/US20100210827A1/en
Publication of KR20090025481A publication Critical patent/KR20090025481A/en
Application granted granted Critical
Publication of KR100890186B1 publication Critical patent/KR100890186B1/en
Priority to US13/108,518 priority patent/US20110281300A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

A method for manufacturing a recombinant protein using RpoS(RNA polymerase sigma factor) is provided to improve yield of the recombinant protein due to a small size of the RpoS. A method for manufacturing a recombinant protein using RpoS(RNA polymerase sigma factor) comprises the following steps of: manufacturing an expression vector by connecting a heterologous protein to a RpoS gene as a fusion partner; manufacturing a transformant by employing the expression vector to a host cell; and inducing expression of the recombinant protein by cultivating the transformant.

Description

RpoS를 융합파트너로 이용한 재조합 단백질의 제조방법{PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER}Production method of recombinant protein using RSP as a fusion partner {PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER}

본 발명은 RpoS(RNA polymerase sigma factor)를 융합파트너로 이용하는 재조합 단백질의 발현벡터 및 이를 이용한 재조합 단백질을 제조하는 방법에 관한 것으로, 더욱 상세하게는 융합파트너(fusion partner) 및 외래단백질의 유전자를 연결한 발현벡터를 제조하고, 상기 발현벡터를 숙주세포에 도입하여 형질전환체를 제조하고, 상기 형질전환체를 배양하여 재조합 단백질의 발현을 유도하고 이를 수득하는 공정을 포함하는 재조합 단백질의 생산방법에 있어서, RpoS의 유전자를 융합파트너로 사용하여 발현시킴으로써 재조합 단백질의 수용성 및 접힘(folding)을 향상시키는 것을 특징으로 하는 재조합 단백질의 제조방법 및 RpoS의 유전자를 융합파트너로 포함된 발현벡터에 관한 것이다.The present invention relates to an expression vector of a recombinant protein using RpoS (RNA polymerase sigma factor) as a fusion partner, and a method for producing a recombinant protein using the same, and more particularly, to connect a fusion partner and a gene of a foreign protein. A method of producing a recombinant protein comprising the steps of preparing an expression vector, introducing the expression vector into a host cell to produce a transformant, and culturing the transformant to induce expression of and obtain the recombinant protein. The present invention relates to a method for producing a recombinant protein, characterized in that to improve the water solubility and folding (folding) of the recombinant protein by using the expression of the RpoS gene as a fusion partner, and to an expression vector containing the gene of RpoS as a fusion partner.

현재 생명공학 기술에 의해 생산되는 단백질에는 일반적으로 면역 조절 및 효소 저해제 및 호르몬 같은 의약 및 연구용 단백질과 진단용 단백질이나 반응 첨가 효소와 같은 산업용 단백질로 대별될 수 있으며 이 두 가지 단백질들을 중심으 로 생산 공정 기술 개발 및 산업화가 추진되고 있다. 특히 재조합 미생물 기술을 이용하여 유용한 재조합 단백질을 생산할 때, 유전정보가 잘 알려져 있으며 다양한 벡터 시스템을 구축하고 있고, 비교적 값싼 배지에서 빠르게 고농도로 배양할 수 있는 장점을 가진 대장균이 연구 또는 상업적 목적으로 많이 사용되고 있다.Currently, proteins produced by biotechnology can be roughly divided into pharmaceutical and research proteins such as immunomodulatory and enzyme inhibitors and hormones, and industrial proteins such as diagnostic proteins and reactive enzymes. Technology development and industrialization are being promoted. In particular, when producing useful recombinant proteins using recombinant microbial technology, E. coli is well known for its genetic information, has a variety of vector systems, and has the advantage of being able to cultivate rapidly at high concentrations in relatively inexpensive media. It is used.

대장균에서 재조합 단백질을 생산함에 있어 강력한 유도성(inducible) 프로모터를 갖춘 다양한 발현벡터가 개발되어 외래단백질의 생산에 이용되어 왔다. 그러나 숙주세포로 대장균을 이용하는 경우 제조하고자 하는 단백질이 대장균 내의 단백질 분해효소에 의해 분해되어 수율이 낮아지는 경우가 많으며, 특히 분자량이 10kDa 이하의 작은 크기의 폴리펩타이드의 발현에서 이러한 경향이 심한 것으로 알려져 있다. 뿐만 아니라 재조합 단백질이 대장균에서 과다 발현시 응집체(inclusion body)로 알려진 불용성 응집체를 형성하는 경우가 많다고 알려져 있으며 응집체로 발현된 폴리펩타이드의 경우 접힘(folding) 중간체가 분자 상호간의 다이설파이드 결합(intermolecular disulfide bond) 또는 소수성 상호작용(hydrophobic interaction)에 의해 숙주세포의 다른 단백질 불순물들[샤페론(chaperon), 라이보좀(ribosome), 초기인자 등]과 비선택적으로 결합함으로써 목적 폴리펩타이드의 응집체 내 순도가 떨어지는 단점이 있다. 또한 이렇게 발현된 단백질을 활성형으로 만들기 위해서는 구아니딘-하이드로클로라이드(Guanidine hychloride)나 우레아(urea)같은 변성제를 사용하여 용해시킨 후 희석하는 재접힘(refolding) 과정을 거쳐야 하는데 이때 단백질이 활성형으로 접히지 않는 등 생산 수율이 감소하는 문제점이 있다.(Marston FA, Biochem J 240(1):1-12, 1986).In producing recombinant proteins in Escherichia coli, various expression vectors with powerful inducible promoters have been developed and used for the production of foreign proteins. However, when E. coli is used as a host cell, the protein to be produced is often degraded by proteolytic enzymes in E. coli, and the yield is low. In particular, the expression of a small polypeptide having a molecular weight of 10 kDa or less is known to be severe. have. In addition, recombinant proteins often form insoluble aggregates, known as inclusion bodies, when they are overexpressed in Escherichia coli. In the case of polypeptides expressed as aggregates, the folding intermediates are intermolecular disulfides. Inferior purity in aggregates of the desired polypeptide by non-selective binding to other protein impurities (chaperon, ribosome, early factor, etc.) of the host cell by bond or hydrophobic interaction There are disadvantages. In addition, in order to make the expressed protein active, it must be refolded by dissolving and diluting with a denaturant such as guanidine hychloride or urea. Production yields are reduced (Marston FA, Biochem J 240 (1): 1-12, 1986).

대장균에서 활성형의 재조합 단백질을 고 수율로 얻기 위해 대장균의 단백질 발현속도를 낮추어 목적단백질의 수용도를 높여 저온 발현 실행(Hammarstrom et al., Protein Sci. 11:313-321, 2002), 다양한 프로모터를 사용하거나 유도(induction) 조건의 최적화(Qing et al ., Nat Biotechnol . 22:877-882, 2004) 및 분자 샤페론 또는 단백질 접힘 조절자와 목적단백질의 동시발현(de Marco & De Marco, J Biotechnol . 109:45-52, 2004) 등이 많이 시도되고 있지만, 정제와 목적단백질의 접힘을 돕는 융합파트너와 목적단백질을 융합하여 발현하는 것이 가장 일반적인 방법이다.In order to obtain high yield of active recombinant protein in Escherichia coli, the expression rate of Escherichia coli is lowered to increase the acceptability of the target protein, thereby lowering temperature expression (Hammarstrom et al. , Protein Sci . 11: 313-321, 2002), using various promoters or optimizing induction conditions (Qing et al . , Nat Biotechnol . 22: 877-882, 2004) and co-expression of molecular chaperone or protein folding regulators with protein of interest (de Marco & De Marco, J Biotechnol . 109: 45-52, 2004). Fusion of the partner and the protein of interest is the most common method.

실제로 대장균 내에서 외래단백질의 수용성 높은 발현을 유도하는 여러 융합파트너가 지난 수년 동안 연구되고 보고되어왔다(Esposito & Chatterjee, Curr Opin Biotechnol . 17:353-358 2006; Kapust & Waugh, Protein Sci . 8:1668-1674, 1999; Sachdev & Chirgwin, Biochem . Biophys . Res . Commun . 244:933-937, 1998). 수많은 융합파트너가 개발되고 이용되어왔지만 가장 많이 연구된 대표적인 융합파트너로는 말토오즈 결합 단백질(Maltose binding protein; MBP), 글루타치온-S-전이효소(Glutathione-S-transferase; GST), 티오레독신(Thioredoxin; Trx), NusA 등이 있다. 말토오스 결합 단백질의 경우 이량체 형성에 관여하는 부분에 소수성을 띠는 아미노산이 모여 만들어진 넓은 소수성 틈새가 새로 합성되는 단백질의 소수성 부위를 효과적으로 감춰주어 목적단백질이 불용성 응집체가 되는 것을 방지해 주며, 티오레독신은 목적단백질의 다이설파이드 결합을 도와주고, NusA의 경우 대장균에서 과량으로 발현되었을 때 활성형으로 접히는 능력이 매우 뛰어나므로 뒤따 라 발현되는 목적단백질의 올바른 접힘을 유도한다(Bach H et al ., J Mol Biol 312:79-93, 2001; Edward RL et al ., Nat Biotechnol 11:187-193, 1993; Davis GD et al ., Biotechnol Bioeng 65:382-388, 1999)Indeed, several fusion partners have been studied and reported over the past several years that induce high water-soluble expression of foreign proteins in Escherichia coli (Esposito & Chatterjee, Curr Opin). Biotechnol . 17: 353-358 2006; Kapust & Waugh, Protein Sci . 8: 1668-1674, 1999; Sachdev & Chirgwin, Biochem . Biophys . Res . Commun . 244: 933-937, 1998). Numerous fusion partners have been developed and used, but the most studied fusion partners are maltose binding protein (MBP), glutathione-S-transferase (GST) and thioredoxin ( Thioredoxin (Trx), NusA, and the like. In the case of maltose-binding protein, a large hydrophobic gap, in which hydrophobic amino acids are gathered in the part involved in dimer formation, effectively hides the hydrophobic portion of the newly synthesized protein, thereby preventing the target protein from becoming an insoluble aggregate. Toxins help disulfide binding of the protein of interest, and NusA has a very good ability to fold into the active form when overexpressed in Escherichia coli, leading to the correct folding of the protein of interest (Bach H et. al . , J Mol Biol 312: 79-93, 2001; Edward RL et al . , Nat Biotechnol 11: 187-193, 1993; Davis GD et al . , Biotechnol Bioeng 65: 382-388, 1999)

이러한 융합파트너는 친화 크로마토그래피(affinity chromatography) 방법을 이용하여 융합발현된 목적단백질을 쉽게 정제할 수 있다는 장점이 있으며 각기 다른 분자생물학적 특성을 이용하여 목적단백질의 접힘을 돕는다고 알려져 있다. 하지만 이러한 융합파트너들은 목적단백질에 비해 상대적으로 크기가 커서 융합 부분의 크기에 따라 목적단백질의 수율이 현저히 떨어진다는 단점과 의료목적 또는 산업적으로 유용한 단백질들에 모두 범용적으로 작용하지 않는다는 단점이 있다. 또한, 목적단백질을 수용성으로의 발현을 유도하더라도 고유한 기능을 수행하는 활성형으로의 발현을 유도하는 데는 실패하는 경우가 많고, 적합한 용도로 이용되기 위하여 융합파트너의 제거 과정이 추가되어야 하는 공정상의 불합리성을 지닌다는 문제점이 있다.Such a fusion partner has the advantage of easily purifying the target protein expressed by affinity chromatography and is known to help fold the target protein using different molecular biological properties. However, these fusion partners have a disadvantage in that the yield of the target protein is considerably decreased according to the size of the fusion moiety, since it is relatively large compared to the target protein, and it does not have a general purpose for both the medical and industrially useful proteins. In addition, even when inducing the expression of the target protein in water-soluble, it often fails to induce the expression of the active form that performs its own function, and in the process of removing the fusion partner in order to be used for proper use There is a problem of irrationality.

지금까지 생명공학 기술에 의해 산업적으로 생산된 국내의 재조합 단백질은 발현 가능한 재조합 단백질들의 생산 공정개발에 치중되어 있어서 핵심 원천 기술인 발현시스템 개발은 외국에 비해 상대적으로 모방 또는 개량 수준의 부가기술로서 개발되고 있는 상태이며 상업적으로나 의약적으로 매우 중요한 단백질은 분비 단백질(secretory protein) 및 막 단백질(membrane protein)들로서 대장균 내에서 발현시 불용성 응집체를 만드는 난발현성 단백질(difficult-to-express proteins)이여서 개발이 지연되고 있다. 이를 극복하기 위해서는 대장균 내에서 난발현 단 백질의 발현시스템에 관한 원천 기반기술을 확보하는 것이 중요하다.Until now, domestic recombinant protein produced industrially by biotechnology has been focused on the development of the production process of expressible recombinant proteins, so the development of expression system, which is a core source technology, is developed as an additional technology of imitation or improvement level compared to other countries. The proteins that are present and are of great commercial and medicinal importance are secretory and membrane proteins, which are difficult-to-express proteins that produce insoluble aggregates when expressed in Escherichia coli. It is becoming. In order to overcome this, it is important to secure a source-based technology for the expression system of E. coli protein in E. coli.

또한 상기한 바와 같이 각각의 융합파트너는 각기 다른 분자생물학적 특성을 가지므로 융합된 재조합 단백질이 접힘을 돕는 기작도 다르기 때문에 모든 단백질에서 동일한 효과를 나타내는 것은 아니다. 좀 더 범용적으로 재조합 단백질의 접힘을 효과적으로 돕는 융합파트너를 이용한 발현 시스템을 구축하기 위해서는 기존에 융합파트너가 가지는 특징을 벗어나 다른 관점에서 융합파트너를 발굴하여 새로운 개념의 융합파트너 라이브러리를 구축해야 한다.In addition, as described above, since each fusion partner has different molecular biological characteristics, the fusion recombinant protein may have different mechanisms to assist folding, so that not all proteins have the same effect. In order to build an expression system using a fusion partner that helps to fold a recombinant protein more universally, it is necessary to find a fusion partner from a different point of view and build a new concept of a fusion partner library from a different point of view.

대장균에서의 재조합 단백질 과다 발현은 외부 환경으로부터 받는 스트레스(열 충격, 아미노산 고갈 등)와 비슷한 영향을 주는 것이 이전의 보고에서 알려진바 있다(Hoffman F & Rinas U, Adv Biochem Eng Biotechnol, 89:73-92, 2004). 이에 본 발명자들은 상기 보고를 감안하여 새로운 개념의 융합파트너를 개발하고자 하였다. 대장균에 단백질의 올바른 접힘을 저해하는 스트레스를 주고, 과발현되는 단백질을 융합파트너로 이용하여 다양한 목적단백질을 유전자 재조합 기술을 이용해 생산한 결과, 다양한 목적단백질에 대해서 수용성 발현량이 많이 증가하고 효소 활성에 변화가 없는 재조합 단백질의 생산이 가능함으로 확인함으로써 본 발명을 완성하였다.Recombinant protein overexpression in Escherichia coli has been known in previous reports to have a similar effect on stress from the external environment (heat shock, amino acid depletion, etc.) (Hoffman F & Rinas U, Adv). Biochem Eng Biotechnol , 89: 73-92, 2004). In view of the above report, the present inventors have attempted to develop a new concept of fusion partner. As a result of stress that inhibits the correct folding of E. coli and the overexpressed protein as a fusion partner, various target proteins were produced by genetic recombination technology, resulting in a large increase in water soluble expression for various target proteins and changes in enzyme activity. The present invention has been completed by confirming that the production of recombinant protein is possible.

본 발명의 목적은 형질전환 미생물로부터 목적단백질을 불용성 응집체의 형태로 생산하는 데 있어, 불용성 응집체를 변성제나 환원제를 사용하지 않고도 활성 단백질로 분리해 내는 동시에 다양한 목적단백질에서 활용 가능한 범용성 융합파트너를 이용하는 재조합 단백질을 제조하는 방법을 제공하는 것이다.An object of the present invention is to produce a protein of interest in the form of an insoluble aggregate from a transforming microorganism, by using a universal fusion partner that can be used in various target proteins while separating the insoluble aggregate into active proteins without using denaturing or reducing agents. It is to provide a method for producing a recombinant protein.

상기 목적을 달성하기 위하여, 본 발명은 1) 융합파트너(fusion partner)로서 RpoS(RNA polymerase sigma factor)의 유전자 및 외래단백질의 유전자를 연결한 발현벡터를 제조하는 단계;In order to achieve the above object, the present invention comprises the steps of: 1) preparing an expression vector connecting the gene of RpoS (RNA polymerase sigma factor) and the gene of the foreign protein as a fusion partner;

2) 상기 발현벡터를 숙주세포에 도입하여 형질전환체를 제조하는 단계; 및,2) preparing a transformant by introducing the expression vector into a host cell; And,

3) 상기 형질전환체를 배양하여 재조합 단백질의 발현을 유도하고 이를 수득하는 단계로 구성되는 재조합 단백질의 제조방법을 제공한다.3) culturing the transformant to induce the expression of the recombinant protein and to provide a method for producing a recombinant protein consisting of obtaining the same.

또한, 본 발명은 융합파트너와 외래단백질의 유전자가 연결되어 포함되어 재조합 단백질을 생산하는 발현벡터에 있어서, 상기 융합파트너는 RpoS의 유전자인 것을 특징으로 하는 발현벡터를 제공한다.In addition, the present invention provides an expression vector, characterized in that the fusion partner is a gene of RpoS in the expression vector that is linked to the gene of the foreign protein is linked to produce a recombinant protein.

또한, 본 발명의 발현벡터가 형질 도입된 형질전환체를 제공한다.The present invention also provides a transformant transformed with the expression vector of the present invention.

아울러, 본 발명의 방법에 의해 제조된 재조합 융합 단백질을 제공한다.In addition, there is provided a recombinant fusion protein produced by the method of the present invention.

이하, 본 발명에서 사용한 용어를 설명한다.Hereinafter, the term used by this invention is demonstrated.

"외래단백질(heterologous protein) 또는 목적단백질(target protein)"은 당업자가 대량으로 생산하고자 하는 단백질로서, 재조합 발현벡터에 상기 단백질을 코딩하는 폴리뉴클레오티드를 삽입하여 형질전환체에서 발현이 가능한 모든 단백질을 의미한다."Heterologous protein or target protein" is a protein that a person skilled in the art intends to produce in large quantities, and inserts a polynucleotide encoding the protein into a recombinant expression vector to express all proteins that can be expressed in a transformant. it means.

"재조합 단백질(recombinant protein) 또는 융합단백질(fusion protien)"은 원래의 외래단백질의 서열의 N-말단 또는 C-말단에 다른 단백질이 연결되거나 다른 아미노산 서열이 부가된 단백질을 의미한다."Recombinant protein or fusion protein" refers to a protein in which another protein is linked to or added to the N-terminus or C-terminus of the sequence of the original foreign protein.

"발현벡터"는 발현벡터의 전사에 제공되는 추가단편에 작동가능하게 연결된 관심의 폴리펩티드를 암호화하는 단편으로 구성되는 선형 또는 원형의 DNA 분자이다. 그와 같은 추가단편은 프로모터 및 종료암호 서열을 포함한다. 발현벡터는 하나 이상의 복제 개시점, 하나 이상의 선택마커, 폴리아데닐화 신호 등을 또한 포함한다. 발현벡터는 일반적으로 플라스미드 또는 바이러스 DNA로부터 유도되거나, 또는 둘 다의 요소를 함유한다.An "expression vector" is a linear or circular DNA molecule consisting of fragments encoding a polypeptide of interest operably linked to additional fragments provided for transcription of the expression vector. Such additional fragments include promoter and termination code sequences. Expression vectors also include one or more replication initiation points, one or more selection markers, polyadenylation signals, and the like. Expression vectors are generally derived from plasmid or viral DNA, or contain elements of both.

이하, 본 발명을 상세히 설명한다.Hereinafter, the present invention will be described in detail.

본 발명은 1) 융합파트너(fusion partner)로서 RpoS의 유전자 및 외래단백질의 유전자를 연결한 발현벡터를 제조하는 단계;The present invention 1) preparing an expression vector connecting the gene of RpoS and the gene of the foreign protein as a fusion partner (fusion partner);

2) 상기 발현벡터를 숙주세포에 도입하여 형질전환체를 제조하는 단계; 및,2) preparing a transformant by introducing the expression vector into a host cell; And,

3) 상기 형질전환체를 배양하여 재조합 단백질의 발현을 유도하고 이를 수득하는 단계로 구성되는 재조합 단백질의 제조방법을 제공한다.3) culturing the transformant to induce the expression of the recombinant protein and to provide a method for producing a recombinant protein consisting of obtaining the same.

대장균에서 외래단백질은 불용성 응집체로 발현되는 경우가 많다. 이에 본 발명에서는 단백질의 올바른 접힘을 저해하는 스트레스를 주어졌을 때 발현이 증가하는 대장균 단백질인 RpoS를 융합파트너로 사용하여, 재조합 단백질의 수용성 발현을 향상시키고자 하였다. 단백질의 접힘을 저해하는 조건에서도 활성이 유지되며 발현량이 증가한 단백질은 단백질의 잘못된 접힘(misfolding)을 극복하기 위한 대장균의 생체 메커니즘에 관여한다는 가능성뿐 아니라 단백질 구조가 안정적이며 자체 접힘 능력이 뛰어나다는 것을 보여주므로 융합단백질로서 효과적으로 이용될 수 있다.In Escherichia coli, foreign proteins are often expressed as insoluble aggregates. Therefore, the present invention was intended to improve the water-soluble expression of recombinant proteins using RpoS, an E. coli protein whose expression increases when given stress that inhibits correct folding of the protein, as a fusion partner. The protein remains active even under conditions that inhibit the folding of the protein, and the increased expression level indicates that the protein structure is stable and has excellent self-folding ability as well as the possibility of being involved in the biomechanism of E. coli to overcome the misfolding of the protein. As shown, it can be effectively used as a fusion protein.

구체적으로, 단백질 분해 효과를 나타내는 GdnHCl을 첨가하여 단백질의 올바른 접힘을 저해하는 스트레스를 주어졌을 때 발현이 증가하는 대장균 단백질을 선별하였으며(도 1 참조), 상기 단백질을 분석·동정한 결과 대장균 RpoS(RNA polymerase sigma factor)의 발현량이 증가한 것을 확인하였다(표 1 참조). 단백질 분해 스트레스에 대항하여 수용성 발현이 증가되는 단백질을 융합파트너로 이용할 경우, 목적단백질의 수용성 발현을 높게 유지시켜줄 것이다. 대장균이 성장을 유지할 수 있는 농도로 배지에 GdnHCl을 첨가한 후 대장균의 단백질체의 변화를 비교하기 위하여 2차원 전기영동을 한 후 발현량이 증가한 단백질을 분석한 결과 특정 단백질의 발현량이 약 6배 증가한 것을 확인하였다(도 1 참조). 상기 발현량 이 증가한 단백질을 매트릭스보조 탈착이온화 질량분석기(Matrix-Assisted Laser Desorption/Ionization - Time Of Flight; MALDI-TOF)-MS를 이용하여 분석·동정한 결과 RpoS임을 확인하였다(표 1 참조).Specifically, E. coli protein with increased expression when GdnHCl, which shows a proteolytic effect, was given stress to inhibit correct folding of the protein was selected (see FIG. 1). As a result of analyzing and identifying the protein, E. coli RpoS ( It was confirmed that the expression level of RNA polymerase sigma factor was increased (see Table 1). When a protein having increased water-soluble expression against proteolytic stress is used as a fusion partner, it will maintain high water-soluble expression of the protein of interest. When GdnHCl was added to the medium to maintain the growth of E. coli, two-dimensional electrophoresis was performed to compare the changes in the protein body of E. coli. It was confirmed (see FIG. 1). Proteins with increased expression levels were analyzed and identified using Matrix-Assisted Laser Desorption / Ionization-Time Of Flight (MALDI-TOF) -MS to confirm that they were RpoS (see Table 1).

상기 외래단백질은 당업자가 원하는 모든 단백질이 가능하며, 특히, 의료, 연구용 및 산업용 단백질, 예를 들어, 항원, 항체, 세포수용체, 효소, 구조 단백질, 혈청, 세포 단백질로 이루어진 군으로부터 선택되는 생물학적 활성을 갖는 다양한 외래단백질을 재조합 단백질로 발현할 수 있다. 본 발명에서는 RpoS가 외래단백질의 접힘을 돕는 융합파트너로서의 가능성을 검증하기 위하여 대장균에서 융합파트너 없이 단독으로 발현하였을 때 불용성 응집체를 형성한다고 알려진 10개의 단백질과 융합 발현하였다. 즉, 의료, 연구용 단백질로는 대표적으로 인간 미니프로인슐린(human minipro-insulin, 이하 mp-INS), 인간 상피세포 성장인자(human epidermal growth factor, 이하 EGF), 인간 프리프로-그렐린(human prepro-ghrelin, 이하 ppGRN), 인간 인터루킨-2(human interleukin-2, 이하 hIL-2), 인간 활성유도 시티딘 디아미네이즈(human activation induced cytidine deaminase, 이하 AID), 인간 글루타메이트 디카르복실레이즈(human glutamate decarboxylase, 이하 GAD448 -585), 수도모나스 푸티다(Pseudomonas putida) 유래의 큐티네이즈(cutinase, 이하 CUT), 인간 페리틴 가벼운 사슬(human ferritin light chain, 이하 hFTN-L), 인간 과립구 집락-자극인자(human granulocyte colony-stimulating factor, 이하 G-CSF) 및 한냉 자가염증 증후군 Nacht 도메인[cold autoinflammatory syndrome1(NALP3) Nacht domain, 이하 Nacht]을 목적단백질로 융 합 발현하였고 산업용 단백질로는 제1형 당뇨병의 진단 마커로 알려진 글루탐산 탈카르복시화 효소의 448 내지 558 아미노산(glutamate decarboxylase448-585, 이하 'GAD448-585')과 섬유의 전처리에 이용되고 플라스틱 분해능을 가져 친환경 산업용 효소로 각광받고 있는 수도모나스 푸티다(Pseudomonas putida) 유래의 큐티네이즈(cutinase, 이하 'CUT')를 목적단백질로 선정하여 각각 RpoS의 카르복실 말단에 삽입한 발현 벡터를 제조한 후(도 2 참조) 이를 대장균에 형질전환시키고 이로부터 재조합 단백질 형태로 생산하였다. 각각의 재조합 단백질의 발현량을 확인한 결과, RpoS와 융합한 외래단백질의 발현이 대부분 불용성 응집체보다 수용성 발현에서 더 많은 것을 확인하였고(도 3 참조), 또한 단독 발현한 외래단백질의 수용성 발현량보다 RpoS와 융합한 외래단백질에서 수용성 발현량이 매우 증가하는 것을 확인하였다(도 4 참조).The foreign protein may be any protein desired by those skilled in the art, and in particular, a biological activity selected from the group consisting of medical, research and industrial proteins such as antigens, antibodies, cell receptors, enzymes, structural proteins, serum, and cellular proteins. Various foreign proteins with can be expressed as recombinant proteins. In the present invention, RpoS was fused and expressed with 10 proteins known to form insoluble aggregates when expressed alone without a fusion partner in E. coli to verify the potential as a fusion partner to help fold foreign proteins. That is, medical and research proteins typically include human minipro-insulin (MP-INS), human epidermal growth factor (EGF), human prepro-grelin (human prepro-). ghrelin (ppGRN), human interleukin-2 (hIL-2), human activation induced cytidine deaminase (AID), human glutamate decarboxylase, hereinafter GAD 448 -585), also the Pseudomonas footage (Pseudomonas putida ) cutinase (CUT), human ferritin light chain (hFTN-L), human granulocyte colony-stimulating factor (G-CSF) and cold Cold autoinflammatory syndrome (NALP3) Nacht domain (hereinafter referred to as Nacht) was fusion-expressed as the target protein, and industrial proteins were 448 to 558 amino acids of glutamic acid decarboxylase known as diagnostic markers of type 1 diabetes. glutamate decarboxylase 448-585, hereinafter 'GAD 448-585') and is used in the pretreatment of fiber may also receiving attention as environmentally friendly industrial enzyme brought plastic resolution footage is Pseudomonas (Pseudomonas putida ) -derived cutinase (hereinafter referred to as 'CUT') was selected as the target protein, respectively, to prepare expression vectors inserted into the carboxyl ends of RpoS (see FIG. 2), and then transformed into E. coli and recombinant proteins therefrom. Produced in the form. As a result of confirming the expression level of each recombinant protein, it was confirmed that the expression of the foreign protein fused with RpoS was more in the water-soluble expression than the insoluble aggregate (see FIG. 3), and also the RpoS than the water-soluble expression of the foreign protein expressed alone. It was confirmed that the amount of water-soluble expression in the foreign protein fused with and very increased (see Fig. 4).

또한, 상기의 방법으로 재조합 단백질의 활성을 검증하기 위해 융합발현 파트너가 따로 제거되지 않아도 사용할 수 있는 산업용 단백질인 재조합 큐티네이즈(RpoS::CUT)의 PNP 특이적 분해 활성을 확인하였다(도 5 참조).In addition, PNP specific degradation activity of recombinant cutinase (RpoS :: CUT), which is an industrial protein that can be used without separately removing a fusion expression partner, was verified to verify the activity of the recombinant protein by the above method (see FIG. 5). ).

또한, 본 발명은 융합파트너와 외래단백질의 유전자가 연결되어 포함되어 재조합 단백질을 생산하는 발현벡터에 있어서, 상기 융합파트너는 RpoS의 유전자인 것을 특징으로 하는 발현벡터를 제공한다.In addition, the present invention provides an expression vector, characterized in that the fusion partner is a gene of RpoS in the expression vector that is linked to the gene of the foreign protein is linked to produce a recombinant protein.

뼈대 벡터에 융합 파트너로서 RpoS 유전자와 외래단백질의 유전자가 연결되 어 포함되며, 본 발명의 방법에서 사용될 수 있는 뼈대 벡터는 특별히 이에 제한되는 것은 아니나, pT7, pET/Rb, pGEX, pET28a, pET-22b(+) 및 pGEX로 이루어진 군으로부터 선택되는 대장균에 형질전환 가능한 다양한 벡터를 사용할 수 있다. 본 발명의 상기 발현벡터는 pT7 뼈대 벡터에 상기 융합파트너의 유전자와 외래단백질의 유전자를 삽입할 부위를 일련의 순서로 포함함으로써 상기 융합파트너와 외래단백질을 재조합 단백질로 발현할 수 있다(도 2 참조).The RpoS gene and the foreign protein gene are linked to the skeletal vector as a fusion partner, and the skeletal vector which can be used in the method of the present invention is not particularly limited, but is not limited to pT7, pET / Rb, pGEX, pET28a, pET- Various vectors capable of transforming E. coli selected from the group consisting of 22b (+) and pGEX can be used. The expression vector of the present invention can express the fusion partner and the foreign protein as a recombinant protein by including a sequence of insertion of the gene of the fusion partner and the gene of the foreign protein in the pT7 skeleton vector in a sequence (see FIG. 2). ).

더 나아가, 본 발명의 벡터의 융합파트너의 유전자에 작동 가능하도록 RpoS 유전자와 외래단백질 사이에 단백질 절단효소 인식부위를 코딩하는 폴리뉴클레오티드가 연결될 수 있다. 융합파트너와 융합할 목적단백질이 산업용 단백질일 경우, 융합파트너가 목적단백질의 기능을 저하할 수도 있으며, 의학용 단백질일 경우는 항원항체 반응을 야기할 수 있으므로 융합파트너는 제거되는 것이 바람직하다. 이때, 상기 단백질 절단효소 인식부위는 Xa 인자 인식부위, 엔테로키나제 인식부위, 제네나제(Genenase) I 인식부위 또는 퓨린(Furin) 인식부위가 단독으로 사용되거나 어느 두 개 이상을 순차적으로 연결하여 사용할 수 있다. 발명의 실시태양에서, 본 발명의 벡터는 pET28a 뼈대 벡터에 상기 융합파트너의 유전자, 단백질 절단효소 인식부위 및 외래단백질의 유전자를 삽입할 부위를 일련의 순서로 포함함으로써 상기 융합파트너와 외래단백질을 재조합 단백질로 발현할 수 있다.Furthermore, a polynucleotide encoding a protein cleavage enzyme recognition site may be linked between the RpoS gene and the foreign protein to be operable to the gene of the fusion partner of the vector of the present invention. When the target protein to be fused with the fusion partner is an industrial protein, the fusion partner may decrease the function of the target protein, and when the protein is a medical protein, the fusion partner may be removed because it may cause an antigen-antibody reaction. In this case, the protein cleavage enzyme recognition site may be used as the Xa factor recognition site, the enterokinase recognition site, the Genenase I recognition site, or the Furin recognition site, or any two or more of them are sequentially connected. have. In an embodiment of the invention, the vector of the present invention recombines the fusion partner and the foreign protein by including a sequence of insertion of the gene of the fusion partner, the protein cleavage enzyme recognition site, and the foreign protein gene into the pET28a skeleton vector. Can be expressed as a protein.

발명의 실시태양에서, 상기 외래 유전자는 제한효소 부위를 통해 클로닝될 수 있고, 단백질 절단효소 인식부위를 코딩하는 폴리뉴클레오티드가 사용된 경우에는 상기 폴리뉴클레오티드와 틀이 맞도록(in frame) 연결되어, 외래단백질 분비 후 단백질 절단효소로 절단 시, 원래 형태의 외래단백질이 생산될 수 있도록 할 수 있다.In an embodiment of the invention, the foreign gene can be cloned through a restriction enzyme site, and if a polynucleotide encoding a protein cleavage site is used, it is linked in frame with the polynucleotide, When secreted by a protein cleavage enzyme after secreting the foreign protein, the original protein can be produced.

아울러, 본 발명의 벡터의 융합파트너 또는 외래단백질의 유전자에 작동 가능하도록 재조합 단백질의 분리 정제가 용이하도록 분리정제용 태그를 코딩하는 폴리뉴클레오티드가 연결될 수 있다. 이때, 상기 분리정제용 태그는 GST, poly-Arg, FLAG, poly-His 및 c-myc 등이 단독으로 사용되거나 어느 두 개 이상을 순차적으로 연결하여 사용할 수 있다. 발명의 실시태양에서, 본 발명의 벡터는 pT7 뼈대 벡터에 상기 융합파트너의 유전자, 외래단백질의 유전자 및 분리정제용 태그를 삽입할 부위를 일련의 순서로 포함함으로써 상기 융합파트너, 외래단백질 및 분리정제용 태그를 포함하는 재조합 단백질로 발현할 수 있다. 발명의 바람직한 실시예에서는 상기 poly-His 태그를 코딩하는 폴리뉴클레오티드가 연결되었으며, 단백질 발현 및 정제 후 poly-His 태그를 이용하여 재조합 단백질을 정제한 후, SDS-PAGE를 이용하여 재조합 단백질만이 정제된 것을 확인하였고(도 6-1 참조), HPLC로 분석한 결과 단일 피크(peak)가 존재하는 것은 확인하였다(도 6-2 참조). 단백질이 동일한 3차 구조를 형성하고 있으면 HPLC 분석 결과 단일 피크로 나타나기 때문에, 상기 실시예의 결과는 본 발명의 재조합 단백질이 구조적으로 일정하며 동시에 모두 동일한 활성을 나타내는 것을 의미한다.In addition, a polynucleotide encoding a tag for separation and purification may be linked to facilitate separation and purification of a recombinant protein to be operable to a fusion partner or a foreign protein gene of the vector of the present invention. At this time, the separation and purification tag may be used alone, or GST, poly-Arg, FLAG, poly-His and c-myc, or any two or more of them in sequence. In an embodiment of the present invention, the vector of the present invention comprises the fusion partner, the foreign protein, and the separation and purification of the fusion partner by inserting the gene of the fusion partner, the foreign protein gene, and the site for inserting the separation and purification tag in a sequence. It can be expressed as a recombinant protein containing a dragon tag. In a preferred embodiment of the present invention, the polynucleotide encoding the poly-His tag is linked, and after the protein expression and purification, the recombinant protein is purified using a poly-His tag, and only the recombinant protein is purified using SDS-PAGE. It was confirmed that (see Fig. 6-1), HPLC analysis confirmed that the presence of a single peak (peak) (see Fig. 6-2). As the protein forms the same tertiary structure, the HPLC analysis results in a single peak, which means that the recombinant protein of the present invention is structurally constant and simultaneously exhibits the same activity.

또한, 본 발명은 융합파트너와 외래단백질의 유전자가 연결되어 포함되어 재조합 단백질을 생산하는 발현벡터에 있어서, 상기 융합파트너는 RpoS의 유전자인 것을 특징으로 발현벡터가 형질 도입된 형질전환체를 제공한다.In another aspect, the present invention provides a transformant in which the expression vector is transfected, characterized in that the fusion partner and the gene of the foreign protein is linked to produce a recombinant protein, wherein the fusion partner is a gene of RpoS. .

아울러, 본 발명은 1) 융합파트너(fusion partner)로서 RpoS의 유전자 및 외래단백질의 유전자를 연결한 발현벡터를 제조하는 단계;In addition, the present invention 1) preparing an expression vector connecting the gene of RpoS and the gene of the foreign protein as a fusion partner (fusion partner);

2) 상기 발현벡터를 숙주세포에 도입하여 형질전환체를 제조하는 단계; 및,2) preparing a transformant by introducing the expression vector into a host cell; And,

3) 상기 형질전환체를 배양하여 재조합 단백질의 발현을 유도하고 이를 수득하는 단계로 구성되는 재조합 단백질의 제조방법에 의해 제조된 재조합 융합 단백질을 제공한다.3) It provides a recombinant fusion protein produced by the method for producing a recombinant protein consisting of culturing the transformant to induce the expression of the recombinant protein and to obtain it.

본 발명의 재조합 단백질의 융합파트너 RpoS는 크기가 작아서 목적단백질의 수율이 이전의 융합파트너에 비해 훨씬 향상될 수 있다. 또한, 도 3 내지 4에서 나타난 바와 같이 의료목적 또는 산업적으로 유용한 단백질들에 모두 범용적으로 작용할 수 있고, 도 5에 나타난 바와 같이 목적단백질의 수용성 발현을 유도할 뿐만 아니라 고유한 기능을 수행하는 활성형으로의 발현을 유도하는 데 적합하다.The fusion partner RpoS of the recombinant protein of the present invention is small in size, so that the yield of the target protein can be much improved compared to the previous fusion partner. In addition, as shown in Figures 3 to 4 can act universally to all of the medical or industrially useful proteins, as shown in Figure 5 not only induces water-soluble expression of the protein of interest, but also performs an activity that performs a unique function It is suitable for inducing expression in form.

본 발명의 RpoS의 유전자를 융합파트너로 사용하여 재조합 단백질을 생산하는 방법은 기존의 융합파트너가 가지고 있는 수용성과 접힘에 관한 한계를 극복할 수 있고, 단백질 의약품 및 산업용 생산에 폭 넓게 이용될 수 있다.The method for producing a recombinant protein using the gene of RpoS of the present invention as a fusion partner can overcome the limitations on water solubility and folding of existing fusion partners, and can be widely used for protein medicine and industrial production. .

이하, 본 발명을 하기 실시예에 의해 상세히 설명한다.Hereinafter, the present invention will be described in detail by the following examples.

단, 하기 실시예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실시예에 의해 한정되는 것은 아니다.However, the following examples are merely to illustrate the invention, but the content of the present invention is not limited by the following examples.

<< 실시예Example 1> 환경 스트레스 하에 의한 대장균  1> Escherichia coli under environmental stress 단백질체Protein bodies 변화 분석 Change analysis

<1-1> 단백질 분해 스트레스에서 배양된 <1-1> cultured under proteolytic stress 대장균에서In E. coli 발현한 수용성 단백질의 회수 Recovery of expressed water soluble protein

대장균에 단백질의 올바른 접힘을 저해하는 스트레스를 주기 위해 단백질 분해 효과를 나타내는 GdnHCl을 첨가한 배지에서 배양한 대장균에서 발현한 수용성 단백질을 회수하였다.In order to give E. coli a stress that inhibits the correct folding of the protein, water-soluble proteins expressed in E. coli cultured in a medium containing GdnHCl showing a proteolytic effect were recovered.

37℃, 130 rpm 및 1L당 트립톤 10 g, 효모 추출물 10 g, NaCl 5 g으로 조성된 LB 배지에서 배양한 대장균 BL21(Escherichia coli K-12)을 OD600이 0.5에 도달했을 때 새로운 LB 배지(대조군) 및 100 mM GdnHCl을 포함하는 LB 배지(실험군)에서 3시간 더 배양한 후 세포배양액을 6000 rpm 4 ℃에서 원심 분리하여 균체 침전물을 회수하였다. 균체 침전물을 40 mM Tris-HCl 완충액(pH 8.0)으로 두 번 씻어낸 후, 500 ㎕의 파쇄 용액[lysis buffer; 8M Urea, 4%(w/v) CHAPS, 40mM Tris, 단백질 분해효소 제한 혼합물(Protease inhibitor cocktail; Roche Diagnostics GmbH, 독일)]에 현탁한 다음 초음파 파쇄기(Branson sonifier; Branson Ultrasonics Corporation, USA)를 이용하여 파쇄하였다. 파쇄액을 4℃에서 12,000 rpm으로 60분간 원심 분리하여 상등액과 침전물을 분리하여 단백질 응집체를 제거 한 뒤 상등액을 분리하였다. 분리된 상등액의 단백질 농도는 바이오-라드 단백질 분석 키트(Bio-Rad protein assay kit)를 이용하여 측정하였고, 상기 상등액의 수용성 단백질 30 ㎎을 재수화 완충액[rehydration solution, 2 M 싸이오우레아(thiourea), 8 M 우레아(urea), 4%(w/v) 챕스(CHAPS), 1%(w/v) DTT, 1%(w/v) 이동성 양성전해질(carrier ampholyte), pH 4.7]에 녹여 2차원 겔 전기영동(2-dimmensional polyacrylamide gel electrophoresis; 2D-PAGE)용 시료로 -80℃에 냉동보관하였다. Escherichia coli BL21 cultured in LB medium consisting of 37 g, 130 rpm and 10 g of tryptone, 1 g of yeast extract, and 5 g of NaCl coli K-12) was further cultured in fresh LB medium (control) and LB medium containing 100 mM GdnHCl (experimental group) when OD 600 reached 0.5, and then the cell culture was centrifuged at 6000 rpm 4 ° C. The cell precipitate was recovered. After washing the cell precipitate twice with 40 mM Tris-HCl buffer (pH 8.0), 500 μl of crushing solution [lysis buffer; 8M Urea, 4% (w / v) CHAPS, 40 mM Tris, Protease inhibitor cocktail (Roche Diagnostics GmbH, Germany)] and suspended using a Branson sonifier (Branson Ultrasonics Corporation, USA). By crushing. The lysate was centrifuged at 12,000 rpm for 60 minutes at 4 ° C. to separate supernatant and sediment to remove protein aggregates and to separate supernatant. Protein concentration of the isolated supernatant was measured using a Bio-Rad protein assay kit, and 30 mg of the water-soluble protein in the supernatant was rehydrated with a 2 M thiourea. , 8 M urea, 4% (w / v) chaps, 1% (w / v) DTT, 1% (w / v) mobile ampholyte, pH 4.7] 2 Samples for dimensional gel electrophoresis (2-dimmensional polyacrylamide gel electrophoresis; 2D-PAGE) were cryopreserved at -80 ° C.

<1-2> <1-2> 단백질체Protein bodies 분석을 위한 2차원 겔 전기영동 Two-Dimensional Gel Electrophoresis for Analysis

2차원 겔 전기영동을 이용하여 실시예 <2-1>의 방법으로 수득한 대조군과 실험군의 단백질체를 분석하였다.The protein bodies of the control group and the experimental group obtained by the method of Example <2-1> were analyzed using two-dimensional gel electrophoresis.

상기 냉동보관된 단백질을 pI별로 분리하기 위한 1차원 등전점 분리과정은 상기 냉동보관된 단백질을 선형 IPG(immobilized pH gradient) 겔 스트립(pH 4-7, 17 ㎝, ReadyStrip, BIO-RAD, USA)에 45 ㎍ 씩 분주한 뒤, 단백질 IEF 셀 전기영동장치(Bio-Rad Protein IEF cell electrophoresis system, USA)에서 500 V, 2시간; 1000 V, 30분; 2000 V, 30분; 4000 V, 30분; 8000 V, 70000 VHr(Volt-hours) 동안 시행하였다. pI별로 분리된 단백질을 포함하고 있는 상기 IPG 겔 스트립을 1% DTT 및 브로모페놀 블루(bromophenol blue)가 포함된 평형화 용액[epuilibration solution; 50 mM 트리스(Tris), pH 8.6, 6 M 우레아(urea), 30%(v/v) 글리세롤(glycerol), 2% SDS]에서 15분간 반응시킨 뒤, 2.5% 아이오도아세트아마이 드(iodoacetamide)가 포함된 평형화 용액에서 15분간 반응시켰다. 단백질을 분자량에 의해 분리하기 위해 평형화된 겔 스트립을 12.5% 폴리아크릴아마이드 겔을 이용하여 PROTEAN II Xi cell system(Bio-Rad, USA)을 이용하여 2차 전기영동 하였다. 전기영동은 브로모페놀 블루 시약이 겔의 끝 부분에 이를 때까지(30 mA/gel, 12h) 4℃에서 진행하였다.The one-dimensional isoelectric point separation process for separating the cryopreserved protein by pI is performed on the cryopreserved protein in a linear IPG (immobilized pH gradient) gel strip (pH 4-7, 17 cm, ReadyStrip, BIO-RAD, USA). After 45 μg aliquot, 500 V, 2 h in Bio-Rad Protein IEF cell electrophoresis system (USA); 1000 V, 30 minutes; 2000 V, 30 minutes; 4000 V, 30 minutes; 8000 V, 70000 VHr (Volt-hours). The IPG gel strip containing proteins separated by pi was equilibrated with 1% DTT and bromophenol blue [epuilibration solution; 50 mM Tris, pH 8.6, 6 M urea, 30% (v / v) glycerol, 2% SDS] for 15 minutes, followed by 2.5% iodoacetamide. Reaction was carried out in an equilibration solution containing 15 minutes. The gel strips equilibrated to separate proteins by molecular weight were subjected to secondary electrophoresis using a PROTEAN II Xi cell system (Bio-Rad, USA) using 12.5% polyacrylamide gel. Electrophoresis was run at 4 ° C. until bromophenol blue reagent reached the end of the gel (30 mA / gel, 12 h).

Rabilloud 방법(Rabilloud T, Methods Mol Biol, 112:297-305, 1999)에 따라 젤을 은-염색(silver staining)한 후 UMAX 파워룩 1100 스캐너(UMAX, USA)로 스캐닝을 하였고, 이미지 마스터 소프트웨서 버전 4.01(Amersham Biosciences, USA)을 이용하여 젤에 나타난 각 단백질 스팟의 영역당 밀도 변화를 측정, 분석하였다. 상기 분석 결과를 바탕으로, 대조군과 실험군에서 분리한 단백질의 발현량을 비교하였고, 실험군에서 스팟 부피가 6배 증가한 단백질의 스팟을 선정하였다(도 1).Rabilloud T, Methods Mol The gel was silver stained according to Biol , 112: 297-305, 1999) and then scanned with a UMAX PowerLook 1100 scanner (UMAX, USA) and image master software version 4.01 (Amersham Biosciences, USA). The change in density per area of each protein spot shown in the gel was measured and analyzed. Based on the results of the analysis, the expression levels of proteins isolated from the control group and the experimental group were compared, and the spot of the protein with a 6-fold increase in the spot volume was selected from the experimental group (FIG. 1).

<< 실시예Example 2>  2> MALDIMALDI -- TOFTOF -- MSMS 분석을 이용한 환경 스트레스 하에 수용성 발현량이 증가한 대장균 단백질의 동정Identification of Escherichia Coli Protein with Increased Water Soluble Expression Under Environmental Stress Using Analysis

실시예 1에 의해 선정된 단백질 스팟을 추출하여 한국기초과학연구소에 기탁하여, MALDI-TOF(Matrix-Assisted Laser Desorption/Ionization - Time Of Flight, MALD-TOF)-MS분석을 이용하여 동정하였다.The protein spots selected in Example 1 were extracted and deposited in the Korea Basic Science Institute, and identified using MALDI-TOF (Matrix-Assisted Laser Desorption / Ionization-Time Of Flight, MALD-TOF) -MS analysis.

구체적으로, MALDI-TOF-MS 분석을 위해서 실시예 1에 의해 선정된 단백질 스팟을 은-염색된 겔로부터 추출하였다(Gharahdaghi F, et al ., Electrophoresis, 20:601-605, 1999). 추출된 단백질 스팟을 트립신(trypsin, 10.15 ㎎/㎖)을 포함 하는 25 nM 암모늄 바이카보네이트(ammonium bicarbonate, pH 8.0) 용액에서 37℃에서 하룻밤 동안 두어 펩타이드 분해과정을 수행하였다. 분해된 펩타이드는 5%(v/v) TFA, 50% (v/v) ACN 용액을 이용하여 추출하였으며, 상기 추출 과정을 세 번 반복한 후 진공 원심분리기를 이용하여 추출한 펩타이드를 포함하는 용액을 건조시켰다. 건조된 펩타이드를 50% ACN/0.1% TFA 용액에 용해시킨 뒤, 한국기초과학연구소에 기탁하여 MALDI-TOF-MS 시스템(Voyager DE-STR instrument; Biosystems, USA)을 이용하여 분자량을 측정하였다. 측정된 펩타이드의 질량 지문(peptide mass fingerprints)은 Prospector 웹사이트의 MS-FIT (http://prospector.ucsf.edu/prospector/4.0.8/html/msfit.htm)을 이용하여 수행하였으며 단백질 동정을 위한 MS-FIT 데이터베이스로는 Swiss-Prot을 이용하였다. 단백질 동정을 수행한 결과 100 mM GdnHCl 하에서 발현량이 6배 증가한 단백질은 대장균 RpoS로 확인되었다(표 1).Specifically, the protein spots selected by Example 1 for MALDI-TOF-MS analysis were extracted from silver-stained gels (Gharahdaghi F,et al .,Electrophoresis, 20: 601-605, 1999). Peptide digestion was performed by placing the extracted protein spot in a 25 nM ammonium bicarbonate (pH 8.0) solution containing trypsin (10.15 mg / ml) overnight at 37 ° C. The digested peptide was extracted using 5% (v / v) TFA, 50% (v / v) ACN solution, and the solution containing the peptide extracted using a vacuum centrifuge was repeated three times. Dried. The dried peptide was dissolved in a 50% ACN / 0.1% TFA solution, and deposited in the Korea Basic Science Institute to determine the molecular weight using a MALDI-TOF-MS system (Voyager DE-STR instrument; Biosystems, USA). Peptide mass fingerprints were measured using MS-FIT (http://prospector.ucsf.edu/prospector/4.0.8/html/msfit.htm) on the Prospector website. Swiss-Prot was used as the MS-FIT database. As a result of protein identification, the protein with 6-fold increase in expression level under 100 mM GdnHCl was identified as E. coli RpoS (Table 1).

단백질 정보Protein Information 유전자 이름Gene name 유전자 접근 번호a Genetic access number a 단백질 이름Protein name 등전점(pI)/분자량(kDa)Isoelectric point (pI) / molecular weight (kDa) 상대적 발현량 증가d Relative expression increase d 서열 유사성(%)% Sequence Similarity RpoS Rpos P13445 P13445 RNA polymerase sigma factorRNA polymerase sigma factor 이론값b Theoretical value b 실험값c Experimental value c 6 6 18.5 % 18.5% 4.9 / 40.0 4.9 / 40.0 5.2 / 39.0 5.2 / 39.0

a. 유전자 접근 번호 : ExPASy Proteomics Server(http://www.expasy.org/)에서 유전자 정보 검색용 식별번호이다.a. Gene access number: An identification number for genetic information retrieval from ExPASy Proteomics Server (http://www.expasy.org/).

b 이론값은 Compute pI/Mw tool(http://www.expasy.org/tools/pi_tool.html)을 이용하여 수집하였다.b The theoretical values were collected using the Compute pI / Mw tool (http://www.expasy.org/tools/pi_tool.html).

c 실험값은 이차원 전기영동 젤 이미지로부터 산출하였다.c Experimental values were calculated from two-dimensional electrophoretic gel images.

d 대조군 RpoS의 발현량을 1로 하여 실험군 RpoS의 상대적 발현량을 나타내었다.d The expression level of the control group RpoS was set to 1 to indicate the relative expression level of the experimental group RpoS.

<< 실시예Example 3> 아미노 말단에 RpoS를 융합파트너로 포함하는 발현 벡터의 제조 3> Preparation of Expression Vector Comprising RpoS at the Amino Terminal as a Fusion Partner

실시예 1 내지 2의 방법으로 선정된 환경 스트레스 하에 수용성 발현량이 증가한 대장균 단백질 RpoS(RNA polymerase sigma factor)를 융합파트너로 포함하는 발현 벡터를 제조하였다.An expression vector comprising E. coli protein RpoS (RNA polymerase sigma factor) having increased water soluble expression under environmental stress selected by the method of Examples 1 to 2 was prepared as a fusion partner.

RpoS 유전자를 암호화하는 뉴클레오티드를 획득하기 위해, Entrez Nucleotide 데이타베이스의 gi:49175990에서 2864581bp 내지 2865573bp의 서열정보(서열번호 45)를 이용하여 정지 코돈을 제외한 RpoS 유전자의 PCR 증폭을 위한 프라이머쌍을 제작하였다. 또한, 상기 프라이머쌍의 센스 프라이머(서열번호 1: cat atg agt cag aat acg ctg aaa)에는 NdeI 제한효소 인식서열을, 안티센스 프라이머(서열번호 2: ctc gag ctc gcg gaa cag cgc ttc)에는 XhoI 제한효소 인식서열을 포함하도록 제작하였다. PCR은 DNA 중합효소반응용 완충용액(0.25 mM dNTPs; 50 mM KCl; 10 mM (NH4)2SO4; 20 mM Tris-HCl(pH8.8); 2 mM MgSO4; 0.1% Triton X-100)에 대장균으로부터 분리한 염색체를 주형 DNA로 100 ng, 서열번호 1 내지 2로 기재되는 프라이머쌍을 각각 50 pmol을 넣은 다음 Taq DNA 중합효소를 이용하여 수행되었다. 반응 조건은 95℃/30초(변성), 52℃/30초(어닐링), 72℃/60초(신장)로 총 30회 수행하였으며, 그 결과 증폭된 DNA절편의 5' 말단에 NdeI 제한효소 부위와 3' 말단에 XhoI 제한 효소 부위를 포함하는 PCR 산물을 수득하였다. 증폭된 PCR 산물을 제한효소 NdeI과 XhoI으로 각각 처리하여 pT7-7(Novagen, USA)의 제한효소 NdeI과 XhoI자리에 삽입하였고 이를 pT7-RpoS라고 명명하였다.To obtain nucleotides encoding the RpoS gene, primer pairs for PCR amplification of the RpoS gene excluding the stop codon were prepared using sequence information (SEQ ID NO: 45) from 2864581bp to 2865573bp in gi: 49175990 of the Entrez Nucleotide database. . In addition, the sense primer of the primer pair (SEQ ID NO: 1: cat atg agt cag aat acg ctg aaa) has Nde I restriction enzyme recognition sequence, the antisense primer (SEQ ID NO: 2: ctc gag ctc gcg gaa cag cgc ttc), the Xho I It was made to include restriction enzyme recognition sequence. PCR was performed using DNA polymerase buffer solution (0.25 mM dNTPs; 50 mM KCl; 10 mM (NH 4 ) 2 SO 4 ; 20 mM Tris-HCl (pH 8.8); 2 mM MgSO 4 ; 0.1% Triton X-100 Chromosome isolated from Escherichia coli) was added using 100 ng of template DNA and 50 pmol of primer pairs described in SEQ ID NOs: 1 to 2, respectively, followed by Taq DNA polymerase. The reaction conditions were performed 30 times at 95 ° C./30 sec (denatured), 52 ° C./30 sec (annealed), and 72 ° C./60 sec (extension), resulting in Nde I restriction at the 5 ′ end of the amplified DNA fragment. PCR products containing enzyme sites and Xho I restriction enzyme sites at the 3 'end were obtained. The amplified PCR product by restriction enzymes Nde I and Xho I in each treatment was inserted into the restriction enzymes Nde I and Xho I seat of pT7-7 (Novagen, USA) was named this pT7-RpoS.

<< 실시예Example 4> 외래단백질을 융합단백질 형태로 제조하는 발현 벡터 4> Expression vector to prepare foreign protein in the form of fusion protein

RpoS를 융합파트너로 이용하여 다양한 외래단백질의 수용성 발현의 증가를 확인하기 위해 외래단백질의 단독 발현 벡터, 융합파트너와의 융합 발현 벡터 및 단백질 절단효소 인식부위를 포함하는 융합 발현 벡터를 제작하였다.In order to confirm the increase in water-soluble expression of various foreign proteins using RpoS as a fusion partner, a fusion expression vector including a single expression vector of a foreign protein, a fusion expression vector with a fusion partner, and a protein cleavage enzyme recognition site was prepared.

<4-1> 외래단백질 단독 발현 벡터<4-1> foreign protein alone expression vector

인간 미니프로인슐린(human minipro-insulin, 이하 mp-INS; EF518215:1-180bp; 서열번호 46), 인간 상피세포 성장인자(human epidermal growth factor, 이하 EGF; NCBI Nucleotide accession number M15672: 1-165bp), 인간 프리프로-그렐린(human prepro-ghrelin, 이하 ppGRN; NCBI Nucleotide accession number NM;016362: 109-393), 인간 인터루킨-2(human interleukin-2, 이하 hIL-2; NCBI Nucleotide accession number NM;000586: 116-517), 인간 활성유도 시티딘 디아미네이즈(human activation induced cytidine deaminase, 이하 AID; NCBI Nucleotide accession number NM;020661: 77-673bp), 인간 글루타메이트 디카르복실레이즈(human glutamate decarboxylase, 이하 GAD448 -585; 서열번호 24), 수도모나스 푸티다(Pseudomonas putida) 유래의 큐티네이즈(cutinase, 이하 CUT; 서열번호 47), 인간 페리틴 가벼운 사슬(human ferritin light chain, 이하 hFTN-L; NM;000146:200-727bp; 서열번호 48), 인간 과립구 집락-자극인자(human granulocyte colony-stimulating factor, 이하 G-CSF; NCBI Nucleotide accession number NM;172219: 131-655bp) 및 한냉 자가염증 증후군 Nacht 도메인[cold autoinflammatory syndrome1(NALP3) Nacht domain, 이하 Nacht; 서열번호 24] 등의 외래단백질의 5'-말단에 NdeI 제한효소 인식서열과 3'-말단에 HindⅢ 제한효소 인식서열을 포함하도록 제작된 표 2의 프라이머쌍을 각각 50 pmol씩 포함한 DNA 중합효소반응용 완충용액(0.25 mM dNTPs; 50 mM KCl; 10 mM (NH4)2SO4; 20 mM Tris-HCl(pH8.8); 2 mM MgSO4; 0.1% Triton X-100)에 주형 DNA 100 ng을 넣은 다음 Taq DNA 중합효소를 이용하여 95℃/30초(변성), 52℃/30초(어닐링), 72℃/60초(신장)로 총 30회 PCR을 수행하였다.Human minipro-insulin (mp-INS; EF518215: 1-180bp; SEQ ID NO: 46), human epidermal growth factor (EGF; NCBI Nucleotide accession number M15672: 1-165 bp) , Human prepro-ghrelin (ppGRN; NCBI Nucleotide accession number NM; 016362: 109-393), human interleukin-2 (hIL-2; NCBI Nucleotide accession number NM; 000586 116-517), human activation induced cytidine deaminase (AID); NCBI Nucleotide accession number NM; 020661: 77-673bp), human glutamate decarboxylase (GAD) 448-585; SEQ ID NO: 24), also the Pseudomonas footage (Pseudomonas putida ) cutinase (CUT; SEQ ID NO: 47), human ferritin light chain (hFTN-L; NM; 000146: 200-727 bp; SEQ ID NO: 48), human granulocyte colony-stimulation Human granulocyte colony-stimulating factor (G-CSF; NCBI Nucleotide accession number NM; 172219: 131-655 bp) and cold autoinflammatory syndrome Nacht domain, hereinafter Nacht; DNA polymerization comprising 50 pmol each of primer pairs of Table 2 prepared to include Nde I restriction enzyme recognition sequence at 5'-end and 3'-end of Hind III restriction enzyme at 3'-end of foreign protein such as SEQ ID NO. Template DNA in enzyme reaction buffer (0.25 mM dNTPs; 50 mM KCl; 10 mM (NH 4 ) 2 SO 4 ; 20 mM Tris-HCl (pH 8.8); 2 mM MgSO 4 ; 0.1% Triton X-100) 100 ng was added, and then 30 times of PCR were performed using a Taq DNA polymerase at 95 ° C./30 sec (denatured), 52 ° C./30 sec (annealed), and 72 ° C./60 sec (extension).

구체적으로 mp-INS, EGF, ppGRN, hIL-2, AID, CUT, G-CSF 및 hFTN-L은 시작 코돈을 제외하고 정지 코돈을 포함하는 아미노산 서열을 증폭하였고, GAD448 -585의 경우는 448번째 아미노산부터 585번째에 해당하는 138개의 아미노산 서열을(서열번호 23), Nacht는 크라이오파이린(cryopyrin)의 nacht 도메인에 해당하는 316개의 아미노산 서열을 증폭하였다(서열번호 24). 이때, GAD448 -585의 경우는 3'-말단에 ClaI 제한효소 인식서열을 포함하도록 프라이머를 제작하였다.Specifically, mp-INS, EGF, ppGRN, hIL-2, AID, CUT, G-CSF and hFTN-L amplified the amino acid sequence including the stop codon except for the start codon, and 448 for GAD 448-585 . 138 amino acid sequences corresponding to the 585th amino acid (SEQ ID NO: 23), Nacht amplified the 316 amino acid sequence corresponding to the nacht domain of cryopyrin (SEQ ID NO: 24). At this time, in the case of GAD 448 -585 was prepared a primer to include the Cla I restriction enzyme recognition sequence on the 3'-end.

또한, mp-INS, EGF, ppGRN, hIL-2, AID, GAD448 -585, hFTN-L, G-CSF 및 Nacht의 주형 DNA는 상기 단백질들이 많이 발현하는 인간 조직으로부터 RNeasy mini kit(QIAGEN, USA)를 이용하여 전체 RNA를 추출한 뒤, 전체 RNA 1 ㎍과 oligo-d(T) 1 ㎕(Invitrogen, USA, 0.5 ㎍/㎕)에 증류수를 50 ㎕까지 채운 후, AccuPower RT-premix(Bioneer, 한국)에 넣어 RT-PCR(reverse transcription polymerasechain reaction) 반응시켰다. 70℃에서 5분, 4℃에서 5분, 42℃에서 60분, 94℃에서 5분, 4℃에서 5분간 반응하여 cDNA를 합성함으로써 수득하였다. 상기 mp-INS는 인간 췌장 조직, EGF는 상피 세포, ppGRN은 인간 태반, hIL-2, hFTN-L, G-CSF 및 Nacht는 인간 백혈구, AID는 인간 측두엽 세포 및 GAD448 -585는 인간 해마 조직에서 각각 클로닝 하였다. CUT의 경우는 Genomic DNA Purification. kit(promega, USA)을 슈도모나스 푸티다로부터 이용하여 추출한 DNA를 주형 DNA로 사용하였다.In addition, mp-INS, EGF, ppGRN , hIL-2, AID, GAD 448 -585, hFTN-L, G-CSF , and template DNA of Nacht is RNeasy mini kit (QIAGEN, USA from a human tissue that the proteins are expressed much ), Extract total RNA, and fill up to 50 µl of distilled water in 1 µg of total RNA and 1 µl of oligo-d (T) (Invitrogen, USA, 0.5 µg / µl), followed by AccuPower RT-premix (Bioneer, Korea). RT-PCR (reverse transcription polymerasechain reaction) reaction. 5 minutes at 70 ° C, 5 minutes at 4 ° C, 60 minutes at 42 ° C, 5 minutes at 94 ° C, and 5 minutes at 4 ° C to obtain cDNA. The mp-INS is human pancreatic tissue, the epithelial cells EGF, ppGRN is human placenta, hIL-2, hFTN-L , G-CSF is human leukocytes and Nacht, AID is a human cell temporal and GAD 448 -585 of the human hippocampal tissue Cloned from For CUT, Genomic DNA Purification. DNA extracted from Pseudomonas putida was used as template DNA.

프라이머 서열Primer sequence 유전자명Gene name 프라이머primer 서열번호 SEQ ID NO: 염기서열Sequence mp-INSmp-INS 센스 sense 서열번호 3SEQ ID NO: 3 cat atg ttt gtc aac caa cat cat atg ttt gtc aac caa cat 안티센스 Antisense 서열번호 4SEQ ID NO: 4 aag ctt tta gtt aca gta gtt c aag ctt tta gtt aca gta gtt c EGFEGF 센스 sense 서열번호 5SEQ ID NO: 5 cat atg aac tct gac tcc gaa tgc cat atg aac tct gac tcc gaa tgc 안티센스 Antisense 서열번호 6SEQ ID NO: 6 aag ctt tta acg cag ttc cca cca aag ctt tta acg cag ttc cca cca ppGRNppGRN 센스 sense 서열번호 7SEQ ID NO: 7 cat atg ggc tcc agc ttc ctg cat atg ggc tcc agc ttc ctg 안티센스 Antisense 서열번호 8SEQ ID NO: 8 aag ctt tca ctt gtc ggc t aag ctt tca ctt gtc ggc t hIL-2hIL-2 센스 sense 서열번호 9SEQ ID NO: 9 cat atg gca cct act tca agt cat atg gca cct act tca agt 안티센스 Antisense 서열번호 10SEQ ID NO: 10 aag ctt tta tca agt cag tgt aag ctt tta tca agt cag tgt AIDAID 센스 sense 서열번호 11SEQ ID NO: 11 cat atg gac agc ctc ttg atg aac cat atg gac agc ctc ttg atg aac 안티센스 Antisense 서열번호 12SEQ ID NO: 12 aag ctt tca taa caa aag tcc ca aag ctt tca taa caa aag tcc ca GAD448 -585 GAD 448 -585 센스 sense 서열번호 13SEQ ID NO: 13 cat atg cgc cac gtt gat gt cat atg cgc cac gtt gat gt 안티센스 Antisense 서열번호 14SEQ ID NO: 14 atc gat tta taa atc ttg tcc atc gat tta taa atc ttg tcc CUTCUT 센스 sense 서열번호 15SEQ ID NO: 15 cat atg gct ccc ctg ccg gat ac cat atg gct ccc ctg ccg gat ac 안티센스 Antisense 서열번호 16SEQ ID NO: 16 aag ctt tta aag ccc gcg gcg ct aag ctt tta aag ccc gcg gcg ct hFTN-LhFTN-L 센스 sense 서열번호 17SEQ ID NO: 17 cat atg agc tcc cag att cgt cat atg agc tcc cag att cgt 안티센스 Antisense 서열번호 18SEQ ID NO: 18 aag ctt tta gtc gtg ctt gag agt aag ctt tta gtc gtg ctt gag agt G-CSFG-CSF 센스 sense 서열번호 19SEQ ID NO: 19 cat atg act cca ctc gga cct g cat atg act cca ctc gga cct g 안티센스 Antisense 서열번호 20SEQ ID NO: 20 aag ctt tca tgg ctg tgc aag aag ctt tca tgg ctg tgc aag NachtNacht 센스 sense 서열번호 21SEQ ID NO: 21 cat atg act gtg gtg ttc cag cat atg act gtg gtg ttc cag 안티센스 Antisense 서열번호 22SEQ ID NO: 22 aag ctt tca cag cag gta gta c aag ctt tca cag cag gta gta c

상기 PCR 증폭 산물을 NdeI/HindIII(GAD448 -585의 경우는 NdeI/ClaI) 제한 효소로 처리한 후, 대장균 발현벡터 pT7(Novagen, USA)의 NdeI/HindIII(GAD448 -585의 경우는 NdeI/ClaI) 자리에 삽입함으로써 외래단백질의 단독 발현 벡터를 수득하였다.The PCR amplification product Nde I / Hind III (the case of GAD 448 -585 is Nde I / Cla I) was treated with restriction enzymes, Nde I / Hind III of E. coli expression vector pT7 (Novagen, USA) (GAD 448 - In the case of 585 , a single expression vector of a foreign protein was obtained by insertion into the Nde I / Cla I) site.

<4-2> 외래단백질의 융합파트너와의 융합 발현 벡터<4-2> Fusion Expression Vector of a Foreign Protein with a Fusion Partner

mp-INS, EGF, ppGRN, hIL-2, AID, GAD448 -585, CUT, hFTN-L, G-CSF 및 Nacht 등의 외래단백질의 5'-말단에 XhoI 제한효소 인식서열과 3'-말단에 HindⅢ 제한효소 인식서열을 포함하도록 제작된 표 3의 프라이머쌍을 각각 50 pmol씩 포함한 DNA 중합효소반응용 완충용액(0.25 mM dNTPs; 50 mM KCl; 10 mM (NH4)2SO4; 20 mM Tris-HCl(pH8.8); 2 mM MgSO4; 0.1% Triton X-100)에 주형 DNA 100 ng을 넣은 다음 Taq DNA 중합효소를 이용하여 95℃/30초(변성), 52℃/30초(어닐링), 72℃/60초(신장)로 총 30회 PCR을 수행하였다. 구체적으로 hFTN-L, mp-INS, EGF, ppGRN, hIL-2, G-CSF, AID, CUT는 시작 코돈을 제외하고 정지 코돈을 포함하는 아미노산 서열을 증폭하였고, GAD448 -585의 경우는 448번째 아미노산부터 585번째에 해당하는 138개의 아미노산 서열을(서열번호 23), Nacht는 크라이오파이린(cryopyrin)의 nacht 도메인에 해당하는 316개의 아미노산 서열을 증폭하였다(서열번호 24). 이때, GAD448 -585의 경우는 3'-말단에 ClaI 제한효소 인식서열을 포함하도록 프라이머를 제작하였다.mp-INS, EGF, ppGRN, hIL-2, AID, GAD 448 -585, CUT, hFTN-L, Xho I on the 5'-end of the heterologous protein such as G-CSF and Nacht restriction enzyme recognition sequence and a 3 ' DNA polymerase reaction buffer containing 50 pmol of each primer pair of Table 3 prepared to include Hind III restriction enzyme recognition sequence at the end (0.25 mM dNTPs; 50 mM KCl; 10 mM (NH 4 ) 2 SO 4 ; 100 ng of template DNA was added to 20 mM Tris-HCl (pH8.8); 2 mM MgSO 4 ; 0.1% Triton X-100) and then 95 ° C / 30 seconds (denatured) using Taq DNA polymerase, 52 ° C / A total of 30 PCRs were performed at 30 seconds (annealing) and 72 ° C./60 seconds (extension). Specifically, hFTN-L, mp-INS, EGF, ppGRN, hIL-2, G-CSF, AID, and CUT amplified the amino acid sequence including the stop codon except for the start codon, and 448 for GAD 448-585 . 138 amino acid sequences corresponding to the 585th amino acid (SEQ ID NO: 23), Nacht amplified the 316 amino acid sequence corresponding to the nacht domain of cryopyrin (SEQ ID NO: 24). At this time, in the case of GAD 448 -585 was prepared a primer to include the Cla I restriction enzyme recognition sequence on the 3'-end.

프라이머 서열Primer sequence 유전자명Gene name 프라이머primer 서열번호 SEQ ID NO: 염기서열Sequence mp-INSmp-INS 센스 sense 서열번호 25SEQ ID NO: 25 ctc gag ttt gtc aac caa cat ctc gag ttt gtc aac caa cat 안티센스 Antisense 서열번호 26SEQ ID NO: 26 aag ctt tta gtt aca gta gtt c aag ctt tta gtt aca gta gtt c EGFEGF 센스 sense 서열번호 27SEQ ID NO: 27 ctc gag aac tct gac tcc gaa tgc ctc gag aac tct gac tcc gaa tgc 안티센스 Antisense 서열번호 28SEQ ID NO: 28 aag ctt tta acg cag ttc cca cca aag ctt tta acg cag ttc cca cca ppGRNppGRN 센스 sense 서열번호 29SEQ ID NO: 29 ctc gag ggc tcc agc ttc ctg ctc gag ggc tcc agc ttc ctg 안티센스 Antisense 서열번호 30SEQ ID NO: 30 aag ctt tca ctt gtc ggc t aag ctt tca ctt gtc ggc t hIL-2hIL-2 센스 sense 서열번호 31SEQ ID NO: 31 ctc gag gca cct act tca agt ctc gag gca cct act tca agt 안티센스 Antisense 서열번호 32SEQ ID NO: 32 aag ctt tta tca agt cag tgt aag ctt tta tca agt cag tgt AIDAID 센스 sense 서열번호 33SEQ ID NO: 33 ctc gag gac agc ctc ttg atg aac ctc gag gac agc ctc ttg atg aac 안티센스 Antisense 서열번호 34SEQ ID NO: 34 aag ctt tca taa caa aag tcc ca aag ctt tca taa caa aag tcc ca GAD448 -585 GAD 448 -585 센스 sense 서열번호 35SEQ ID NO: 35 ctc gag cgc cac gtt gat gt ctc gag cgc cac gtt gat gt 안티센스 Antisense 서열번호 36SEQ ID NO: 36 atc gat tta taa atc ttg tcc atc gat tta taa atc ttg tcc CUTCUT 센스 sense 서열번호 37SEQ ID NO: 37 ctc gag gct ccc ctg ccg gat ac ctc gag gct ccc ctg ccg gat ac 안티센스 Antisense 서열번호 38SEQ ID NO: 38 aag ctt tta aag ccc gcg gcg ct aag ctt tta aag ccc gcg gcg ct hFTN-LhFTN-L 센스 sense 서열번호 39SEQ ID NO: 39 ctc gag agc tcc cag att cgt ctc gag agc tcc cag att cgt 안티센스 Antisense 서열번호 40SEQ ID NO: 40 aag ctt tta gtc gtg ctt gag agt aag ctt tta gtc gtg ctt gag agt G-CSFG-CSF 센스 sense 서열번호 41SEQ ID NO: 41 ctc gag act cca ctc gga cct g ctc gag act cca ctc gga cct g 안티센스 Antisense 서열번호 42SEQ ID NO: 42 aag ctt tca tgg ctg tgc aag aag ctt tca tgg ctg tgc aag NachtNacht 센스 sense 서열번호 43SEQ ID NO: 43 ctc gag act gtg gtg ttc cag ctc gag act gtg gtg ttc cag 안티센스 Antisense 서열번호 44SEQ ID NO: 44 aag ctt tca cag cag gta gta c aag ctt tca cag cag gta gta c

상기 PCR 증폭 산물을 XhoI/HindIII(GAD448 -585의 경우는 XhoI/ClaI) 제한 효소로 처리한 후, 실시예 3의 방법으로 제작한 발현벡터 pT7-RpoS의 XhoI/HindIII(GAD448-585의 경우는 NdeI/ClaI) 자리에 삽입함으로써 외래단백질의 융합파트너와의 융합 발현 벡터를 수득하였다. 상기의 방법으로 만들어진 플라스미드를 각각 pT7-RpoS::FTN-L, pT7-RpoS::IL-2, pT7-RpoS::EGF, pT7-RpoS::G-CSF, pT7-RpoS::AID, pT7-RpoS::CUT, pT7-RpoS::ppGRN, pT7-RpoS::GAD448-585 및 pT7-RpoS::Nacht라고 명명하였다.The PCR amplification product Xho I / Hind III (the case of GAD 448 -585 are Xho I / Cla I) was treated with restriction enzymes, the expression vector pT7-RpoS Xho of production by the method of Example 3 I / Hind III to give a fusion expression vector for the fusion partner of the foreign protein by inserting a place (in the case of GAD 448-585 is Nde I / Cla I). Plasmids produced by the above method were pT7-RpoS :: FTN-L, pT7-RpoS :: IL-2, pT7-RpoS :: EGF, pT7-RpoS :: G-CSF, pT7-RpoS :: AID, pT7 -RpoS :: CUT, pT7-RpoS :: ppGRN, pT7-RpoS :: GAD448-585 and pT7-RpoS :: Nacht.

<4-3> 6개의 히스티딘을 포함하는 정제용 발현 벡터<4-3> Expression vector for purification comprising 6 histidines

시작 코돈을 제외하고 정지 코돈을 포함하는 외래단백질 CUT의 5'-말단에 XhoI 제한효소 인식서열과 3'-말단에 6개의 히스티딘을 코딩하는 폴리뉴클레오티드 및 HindⅢ 제한효소 인식서열을 포함하도록 센스 프라이머(서열번호 49: ctc gag gct ccc ctg ccg gat ac) 및 안티센스 프라이머(서열번호 50: aag ctt tta gtg atg gtg atg gtg atg aag ccc gcg ct)를 50 pmol씩 포함한 DNA 중합효소반응용 완충용액(0.25 mM dNTPs; 50 mM KCl; 10 mM (NH4)2SO4; 20 mM Tris-HCl(pH8.8); 2 mM MgSO4; 0.1% Triton X-100)에 주형 DNA 100 ng을 넣은 다음 Taq DNA 중합효소를 이용하여 95℃/30초(변성), 52℃/30초(어닐링), 72℃/60초(신장)로 총 30회 PCR을 수행하였다. 구체적으로 시작 코돈을 제외하고 정지 코돈을 포함하는 아미노산 서열을 증폭하였다.Sense to include the Xho I restriction enzyme recognition sequence at the 5'-end of the foreign protein CUT containing the stop codon and the polynucleotide and Hind III restriction enzyme recognition sequences encoding 6 histidines at the 3'-end, except for the start codon. Buffer solution for DNA polymerase reaction containing 50 pmol of primer (SEQ ID NO: 49: ctc gag gct ccc ctg ccg gat ac) and antisense primer (SEQ ID NO: 50: aag ctt tta gtg atg gtg atg gtg atg aag ccc gcg ct) 100 ng of template DNA was added to 0.25 mM dNTPs; 50 mM KCl; 10 mM (NH 4 ) 2 SO 4 ; 20 mM Tris-HCl (pH 8.8); 2 mM MgSO 4 ; 0.1% Triton X-100) PCR was performed a total of 30 times at 95 ° C./30 sec (denatured), 52 ° C./30 sec (annealed), and 72 ° C./60 sec (extension) using DNA polymerase. Specifically, the amino acid sequence including the stop codon was amplified except for the start codon.

CUT의 PCR 산물을 제한효소 XhoI과 HindⅢ으로 각각 처리하여, 실시예 3의 아미노 말단에 RpoS를 융합파트너로 포함하는 발현 벡터의 XhoI과 HindⅢ 자리에 삽입하였고 이를 pT7-RpoS::CUT-His6라고 명명하였다.PCR products of the CUT were treated with restriction enzymes Xho I and Hind III, respectively, and inserted into the Xho I and Hind III sites of the expression vector containing RpoS as a fusion partner at the amino terminus of Example 3, which were then expressed as pT7-RpoS :: CUT. It was named -His6.

<< 실시예Example 5> 재조합 단백질의 수용성 발현 5> Water soluble expression of recombinant protein

실시예 4의 방법으로 제조한 외래단백질의 단독 발현 벡터, 융합파트너와의 융합 발현 벡터 및 6개의 히스티딘을 포함하는 정제용 발현 벡터를 대장균에 형질전환하여 배양한 뒤, IPTG로 재조합 단백질의 발현을 유도함으로써 본 발명의 융합파트너에 의한 수용성 발현의 효과를 확인하였다.E. coli was transformed and cultured with a single expression vector of a foreign protein prepared by the method of Example 4, a fusion expression vector with a fusion partner, and a purified expression vector containing 6 histidines, and then expression of the recombinant protein with IPTG. By induction, the effect of water-soluble expression by the fusion partner of the present invention was confirmed.

<5-1> 대장균 형질전환 및 재조합 단백질의 발현<5-1> Escherichia Coli Transformation and Recombinant Protein Expression

하나한(Hanahan)이 기술한 방법(Hanahan D, DNA Cloning vol.1 109-135, IRS press 1985)에 의해 실시예 4의 벡터들을 대장균에 형질전환 하였다.Method described by Hanahan (Hanahan D, DNA Cloning vol. 1 109-135, IRS press 1985) were transformed into E. coli vectors of Example 4.

구체적으로 CaCl2로 처리한 대장균 BL21(DE3)에 실시예 4의 벡터들을 열충격 방법으로 형질전환시킨 후, 앰피실린(ampicillin)이 포함된 배지에서 배양하여 상기 발현벡터가 형질전환되어 앰피실린 저항성을 나타내는 콜로니를 선별하였다. 융합파트너와의 융합 발현 벡터 pP7-RpoS로 형질전환된 대장균을 BL21(DE3):pP7-RpoS로 명명하였다. 상기 콜로니를 하룻밤 동안 LB 배지에서 배양한 종균 배양액의 일부를 100 ㎎/㎖ 앰피실린을 포함하는 LB 배지에 접종한 다음 37℃에서 200 rpm으로 배양하였다. 배양액의 OD600이 0.5에 이르렀을 때 IPTG를 첨가(1 mM)하여 재조합 유전자의 발현을 유도하였다. IPTG 첨가 후 동일한 조건으로 3 내지 4시간 더 배양하였다.Specifically, the vectors of Example 4 were transformed into E. coli BL21 (DE3) treated with CaCl 2 by a heat shock method, and then cultured in a medium containing ampicillin to transform the expression vector into ampicillin resistance. Representative colonies were selected. E. coli transformed with the fusion expression vector pP7-RpoS with the fusion partner was named BL21 (DE3): pP7-RpoS. A portion of the spawn cultures in which the colonies were cultured in LB medium overnight was inoculated in LB medium containing 100 mg / ml ampicillin and then incubated at 37 ° C. at 200 rpm. When OD 600 of the culture reached 0.5, IPTG was added (1 mM) to induce the expression of recombinant genes. After addition of IPTG, the cells were further incubated for 3 to 4 hours under the same conditions.

<5-2> <5-2> SDSSDS -- PAGEPAGE 를 이용한 가용성으로 생산된 재조합 단백질 확인Identification of Recombinant Protein Produced by Solubility

실시예 5-1의 방법으로 배양한 대장균을 6,000 rpm으로 5분간 원심분리하여 균체 침전물을 회수한 후 5 ㎖의 파쇄 용액(10 mM Tris-HCl 완충액, pH 7.5, 10 mM EDTA)에 현탁하여 초음파 파쇄기(Branson sonifier, Branson Ultrasonics Corporation, USA)를 이용하여 파쇄하였다. 파쇄한 후 13,000 rpm으로 10분간 원심분리한 뒤 상등액과 불용성 응집체를 분리하였다. 분리된 상등액의 단백질 농도는 바이오-라드 단백질 분석 키트(Bio-Rad protein assay kit, USA)를 이용하여 측정하였다. 또한, 상등액과 불용성 응집체를 각각 5×SDS(0.156M Tris-HCl, pH 6.8, 2.5% SDS, 37.5% 글리세롤, 37.5 mM DTT)과 1:4로 섞어 100℃에서 10분간 끓였다. 끓인 시료를 10% SDS-PAGE겔의 웰에 로딩하고 125 V에서 2시간 동안 시료를 전개한 다음 겔을 쿠마시 염색 방법으로 염색한 후 탈색하여 각각의 재조합 단백질의 발현량을 농도계(Densitometer, Bio-Rad, USA)로 확인하고, 수학식 1에 따라 용해도(%)를 계산하였다.E. coli cultured by the method of Example 5-1 was centrifuged at 6,000 rpm for 5 minutes to recover the cell precipitate, and then suspended in 5 ml of crushed solution (10 mM Tris-HCl buffer, pH 7.5, 10 mM EDTA) Crushing was performed using a crusher (Branson sonifier, Branson Ultrasonics Corporation, USA). After crushing, the mixture was centrifuged at 13,000 rpm for 10 minutes, and the supernatant and the insoluble aggregate were separated. Protein concentration of the isolated supernatant was measured using a Bio-Rad protein assay kit (USA). In addition, the supernatant and insoluble aggregates were mixed with 5 × SDS (0.156M Tris-HCl, pH 6.8, 2.5% SDS, 37.5% glycerol, 37.5 mM DTT), respectively, 1: 4 and boiled at 100 ° C. for 10 minutes. The boiled sample was loaded into a well of 10% SDS-PAGE gel, the sample was developed at 125 V for 2 hours, the gel was stained by Coomassie staining, and then decolorized. -Rad, USA) and the solubility (%) was calculated according to Equation (1).

Figure 112007064792986-PAT00001
Figure 112007064792986-PAT00001

그 결과, RpoS와 융합한 외래단백질의 발현이 대부분 불용성 응집체보다 수용성 발현에서 더 많은 것을 확인하였다(도 3). 또한, 단독 발현한 외래단백질의 수용성 발현량보다 RpoS와 융합한 외래단백질에서 용해도가 매우 증가하는 것을 확인하였다(도 4).As a result, it was confirmed that the expression of foreign proteins fused with RpoS was more in water-soluble expression than insoluble aggregates (FIG. 3). In addition, it was confirmed that the solubility in the foreign protein fused with RpoS is significantly increased than the water-soluble expression of the foreign protein expressed alone (FIG. 4).

<5-3> RpoS와 융합 발현한 <5-3> Fusion with RpoS 큐티네이즈의Cutinase 효소 활성 Enzyme activity

실시예 5-1 내지 5-2를 통해서 확보한 RpoS 융합발현된 큐티네이즈(이하 RpoS::CUT)의 가수분해 활성을 측정하였다.The hydrolytic activity of the RpoS fusion-expressed cutinase (hereinafter RpoS :: CUT) obtained through Examples 5-1 to 5-2 was measured.

96-웰 마이크로플레이트에 106.7 ㎕의 0.1M 인산완충액(pH 8.0) 및 13.3 ㎕의 Triton X-100(4 g/L)을 넣은 후, 정제하지 않은 RpoS::CUT 상등액과 대조군으로 대장균 BL21(DE3)의 상등액 13.3 ㎕를 각각 첨가하였다. 상기 용액에 66.7 ㎕의 기질(PNP(p-nitrophenyl palmitate) 및 PNB(p-nitrophenyl butyrate), 6.6 mM; Sigma, USA)을 첨가함으로써 가수분해 반응을 시작하였고, 37℃에서 20분간 반응하였다. 반응이 시작된 후 매분 415 ㎚ 파장에서 흡광도의 변화를 측정하였다(Bio-Rad microplate reader, USA).106.7 μl of 0.1M phosphate buffer (pH 8.0) and 13.3 μl of Triton X-100 (4 g / L) were added to a 96-well microplate, followed by E. coli BL21 (DE3) as an unpurified RpoS :: CUT supernatant and a control. 13.3 μl of supernatant) were added, respectively. The hydrolysis reaction was started by adding 66.7 μl of substrate (p-nitrophenyl palmitate) and PNB (p-nitrophenyl butyrate, 6.6 mM; Sigma, USA) to the solution and reacted at 37 ° C. for 20 minutes. After the reaction was started, the change in absorbance at 415 nm wavelength was measured every minute (Bio-Rad microplate reader, USA).

그 결과 상기 수용성 재조합 단백질 RpoS::CUT이 PNP 특이적 분해 능력을 가짐을 확인하였다(도 5).As a result, it was confirmed that the water-soluble recombinant protein RpoS :: CUT has PNP-specific degradation capacity (FIG. 5).

<5-4> 히스티딘 태그를 이용한 재조합 단백질의 정제<5-4> Purification of Recombinant Protein Using Histidine Tag

실시예 4-3의 방법으로 제조한 6개의 히스티딘을 포함하는 정제용 발현 벡터를 실시예 5-1의 하나한(Hanahan)이 기술한 방법(Hanahan D, DNA Cloning vol.1 109-135, IRS press 1985)에 의해 대장균 BL21(DE3)에 형질전환한 후, 재조합 유전자의 발현을 유도하였다. 상기 형질전환된 BL21(DE3)을 실시예 5-2의 방법으로 파쇄한 뒤, 원심분리함으로써 상등액과 불용성 응집체를 분리하였다.The method described by Hanahan of Example 5-1 (Hanahan D, DNA ) for a purified expression vector containing six histidines prepared by the method of Example 4-3 Cloning vol. 1 109-135, IRS press 1985) was transformed into E. coli BL21 (DE3), and the expression of recombinant genes was induced. The transformed BL21 (DE3) was disrupted by the method of Example 5-2, and then the supernatant and the insoluble aggregate were separated by centrifugation.

상기 상등액으로부터 친화성 크로마토그래피법에 의해 재조합 단백질 RpoS::CUT-His6를 정제하였다. 구체적으로 금속 이온이 충진되어 있는 ProBond 레진(Ni2 +) 컬럼(Invitrogen, USA)을 결합 완충액[binding buffer; pH 8.0, 50 mM 소 듐 포스페이트(sodium phosphate), 300 mM 염화 나트륩 (NaCl), 10 mM 이미다졸(imidazole)]을 이용하여 세척하였고, 세척된 컬럼에 RpoS::CUT-His6를 포함하는 상등액을 4℃에서 결합시켰다. 이 후, 8 ㎖의 세척 완충액[washing buffer; pH 8.0, 50 mM 소듐 포스페이트(sodium phosphate), 300 mM 염화 나트륨(NaCl), 50 mM 이미다졸(imidazole)]을 이용하여 두 번 세척하였고, 용리 완충액[elution buffer; pH 8.0, 소듐 포스페이트(sodium phosphate), 300 mM 염화 나트륨 (NaCl), 250 mM 이미다졸(imidazole)]을 이용하여 RpoS::CUT-His6를 회수하였다.From the supernatant, recombinant protein RpoS :: CUT-His 6 was purified by affinity chromatography. Specifically ProBond resin that is filled with the metal ions (Ni + 2) column (Invitrogen, USA) with binding buffer [binding buffer; pH 8.0, 50 mM sodium phosphate, 300 mM sodium chloride (NaCl), 10 mM imidazole], and the washed column contains RpoS :: CUT-His 6 . Supernatants were bound at 4 ° C. Thereafter, 8 ml of washing buffer; pH 8.0, 50 mM sodium phosphate, 300 mM sodium chloride (NaCl), 50 mM imidazole] and washed twice with elution buffer; RpoS :: CUT-His 6 was recovered using pH 8.0, sodium phosphate, 300 mM sodium chloride (NaCl), 250 mM imidazole].

RpoS::CUT-His6를 포함하는 용리 완충액(elution buffer) 내에 존재하는 이미다졸(imidazole)을 제거하기 위해 아미콘 울트라-4 원심분리용 필터(Amicon Ultra-4 centrifugal filter, Millipore, USA)를 이용하였고, PBS 완충용액[PBS buffer; 137 mM 염화 나트륩(NaCl), 2.7 mM 염화 칼륨(KCl), 10 mM 제1인산나트륨(Na2HPO4), 2 mM 제1인산칼륨(KH2PO4), pH 7.4]을 이용하여 완충용액 교환을 수행하였다.Amicon Ultra-4 centrifugal filter (Millipore, USA) was used to remove imidazole present in the elution buffer containing RpoS :: CUT-His 6 . PBS buffer [PBS buffer; 137 mM sodium chloride (NaCl), 2.7 mM potassium chloride (KCl), 10 mM sodium monophosphate (Na 2 HPO 4 ), 2 mM potassium monophosphate (KH 2 PO 4 ), pH 7.4] Solution exchange was performed.

분리된 상등액과 정제된 RpoS::CUT-His6를 각각 5×SDS(0.156M Tris-HCl, pH 6.8, 2.5% SDS, 37.5% 글리세롤, 37.5 mM DTT)과 1:4로 섞어 100℃에서 10분간 끓였다. 끓인 시료를 10% SDS-PAGE겔의 웰에 로딩하고 125 V에서 2시간 동안 시료를 전개한 다음 겔을 쿠마시 염색 방법으로 염색한 후 탈색하여 각각의 재조합 단백질의 발현량을 확인하였다.Separate supernatant and purified RpoS :: CUT-His 6 with 5 × SDS (0.156M Tris-HCl, pH 6.8, 2.5% SDS, 37.5% Glycerol, 37.5 mM DTT), respectively, 1: 4 Boiled for a minute. Boiled samples were loaded into wells of a 10% SDS-PAGE gel, the samples were developed at 125 V for 2 hours, and the gels were stained by Coomassie staining and then decolorized to confirm the expression level of each recombinant protein.

또한, 정제된 RpoS::CUT-His6를 HPLC(high-performance liquid chromatography)를 이용하여 분석하였다. 구체적으로 분석 기구로는 LC-20A Prominence 모델(Shimadzu Co. Ltd., 일본)과 Shim-pack CLC-NH2 컬럼(60 × 150 ㎜, Shimadzu Co. Ltd., 일본)을 이용하였고, 상기 컬럼을 이용한 분석 전에 0.1%의 트리플루오로아세트산(trifluoroacetic acids; SIGMA, USA)를 포함하고 있는 10% 아세토나이트릴(acetonitrile; SIGMA, USA)을 1 ㎖/분의 속도로 흘려주어 평형화하였다. 이에 상기 방법으로 정제한 RpoS::CUT-His6를 평형화된 컬럼에 주입한 뒤 5% 아세토나이트릴을 1 ㎖/분의 속도로 15분간 흘려주어 컬럼으로부터 용리하였다. 용리 분포(elution profile)는 215 ㎚ 파장을 이용하여 검출하였다.In addition, purified RpoS :: CUT-His 6 was analyzed using high-performance liquid chromatography (HPLC). Specifically, an LC-20A Prominence model (Shimadzu Co. Ltd., Japan) and a Shim-pack CLC-NH 2 column (60 × 150 mm, Shimadzu Co. Ltd., Japan) were used as analytical instruments. 10% acetonitrile (SIGMA, USA) containing 0.1% of trifluoroacetic acids (SIGMA, USA) was equilibrated by flowing 1 mL / min. Thus, RpoS :: CUT-His 6 purified by the above method was injected into the equilibrated column, and eluted from the column by flowing 5% acetonitrile at a rate of 1 ml / min for 15 minutes. Elution profiles were detected using a 215 nm wavelength.

그 결과, 수용성 상등액으로부터 RpoS::CUT-His6만이 정제된 것을 확인할 수 있었다(도 6-1). HPLC 분석 결과 하나의 피크(peak)가 존재하는 것을 확인하였다(도 6-2).As a result, it was confirmed that only RpoS :: CUT-His 6 was purified from the aqueous supernatant (FIG. 6-1). HPLC analysis confirmed that there was one peak (Fig. 6-2).

도 1은 환경 스트레스 하에 의한 대장균 단백질체 변화를 분석한 결과도이다:1 is a result of analyzing the change of E. coli protein body under environmental stress:

도 A: 2-D PAGE(2-dimensional polyacrylamide gel electrophoresis) 결과A: Results of 2-D PAGE (2-dimensional polyacrylamide gel electrophoresis)

도 B: GdnHCl에 의한 RpoS 발현량의 증가B: Increase of RpoS expression by GdnHCl

도 2는 외래단백질 단독 발현 벡터, 융합파트너와의 융합 발현 벡터 및 단백질 절단효소 인식부위를 포함하는 융합 발현 벡터의 개열지도이다:2 is a cleavage map of a fusion expression vector comprising a foreign protein alone expression vector, a fusion expression vector with a fusion partner, and a protein cleavage enzyme recognition site:

도 A: 단독 발현 벡터의 개열지도A: cleavage map of a single expression vector

도 B: 융합 발현 벡터의 개열지도B: Cleavage map of fusion expression vector

도 C: 6개의 히스티딘을 포함하는 정제용 발현 벡터의 개열지도Fig. C: cleavage map of a purified expression vector comprising six histidines

도 3은 RpoS 융합 발현한 재조합 단백질의 수용성 상등액(S)과 불용성 응집체(IS)에서의 발현량을 비교한 결과도이다.Figure 3 is a result of comparing the expression in the water-soluble supernatant (S) and insoluble aggregate (IS) of the recombinant protein fusion-expressed RpoS.

도 4는 단독 발현 재조합 단백질과 RpoS 융합 발현한 재조합 단백질의 수용성 발현량을 비교한 그래프이다.Figure 4 is a graph comparing the water-soluble expression of the recombinant protein expressed alone and recombinant RpoS fusion expression.

도 5는 큐티네이즈를 발현하지 않은 대장균의 정제되지 않은 수용성 단백질과 RpoS::CUT의 형태로 RpoS 융합 발현한 재조합 단백질을 발현하는 대장균의 정제 되지 않은 수용성 단백질의 PNB(●) 및 PNP(○)의 분해능을 측정한 결과도이다.Figure 5 shows the PNB (●) and PNP (○) of the unrefined water-soluble protein of E. coli, which does not express the cutinase, and the unrefined water-soluble protein of E. coli, which expresses the recombinant protein expressed by RpoS fusion in the form of RpoS :: CUT. The result of measuring the resolution of is shown.

도 6은 His 태그를 이용하여 재조합 단백질을 정제한 결과를 나타낸 도이다:6 is a diagram showing the result of purification of recombinant protein using His tag:

도 A: SDS-PAGE 법을 이용한 정제 결과Figure A: Purification results using the SDS-PAGE method

M: 단백질 마커M: protein marker

1: RpoS::CUT-His6를 포함하는 정제되지 않은 수용성 상등액1: Unrefined aqueous supernatant containing RpoS :: CUT-His6

2와 3: 정제된 RpoS::CUT-His62 and 3: Purified RpoS :: CUT-His6

도 B: HPLC를 이용한 정제 결과B: Purification results using HPLC

<110> KOREA UNIVERSITY Industry & Academy Collaboration Foundation <120> PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER <130> 7P-08-24 <160> 50 <170> KopatentIn 1.71 <210> 1 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> RpoS Sense Primer <400> 1 catatgagtc agaatacgct gaaa 24 <210> 2 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> RpoS Antisense Primer <400> 2 ctcgagctcg cggaacagcg cttc 24 <210> 3 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Sense Primer <400> 3 catatgtttg tcaaccaaca t 21 <210> 4 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Antisense Primer <400> 4 aagcttttag ttacagtagt tc 22 <210> 5 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Sense Primer <400> 5 catatgaact ctgactccga atgc 24 <210> 6 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Antisense Primer <400> 6 aagcttttaa cgcagttccc acca 24 <210> 7 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Sense Primer <400> 7 catatgggct ccagcttcct g 21 <210> 8 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Antisense Primer <400> 8 aagctttcac ttgtcggct 19 <210> 9 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Sense Primer <400> 9 catatggcac ctacttcaag t 21 <210> 10 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Antisense Primer <400> 10 aagcttttat caagtcagtg t 21 <210> 11 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> AID Sense Primer <400> 11 catatggaca gcctcttgat gaac 24 <210> 12 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> AID Antisense Primer <400> 12 aagctttcat aacaaaagtc cca 23 <210> 13 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Sense Primer <400> 13 catatgcgcc acgttgatgt 20 <210> 14 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Antisense Primer <400> 14 atcgatttat aaatcttgtc c 21 <210> 15 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Sense Primer <400> 15 catatggctc ccctgccgga tac 23 <210> 16 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Antisense Primer <400> 16 aagcttttaa agcccgcggc gct 23 <210> 17 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Sense Primer <400> 17 catatgagct cccagattcg t 21 <210> 18 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Antisense Primer <400> 18 aagcttttag tcgtgcttga gagt 24 <210> 19 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Sense Primer <400> 19 catatgactc cactcggacc tg 22 <210> 20 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Antisense Primer <400> 20 aagctttcat ggctgtgcaa g 21 <210> 21 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nacht Sense Primer <400> 21 catatgactg tggtgttcca g 21 <210> 22 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nacht Antisense Primer <400> 22 aagctttcac agcaggtagt ac 22 <210> 23 <211> 948 <212> DNA <213> Human GAD448-585 <400> 23 accgtggtgt tccagggggc ggcagggatt gggaaaacaa tcctggcccg gaagatgatg 60 ttggactggg cgtcggggac actctaccaa gacaggtttg actatctgtt ctatatccac 120 tgtcgggagg tgagccttgt gacacagagg agcctggggg acctgatcat gagctgctgc 180 cccgacccaa acccacccat ccacaagatc gtgagaaaac cctccagaat cctcttcctc 240 atggacggct tcgatgagct gcaaggtgcc tttgacgagc acataggacc gctctgcact 300 gactggcata aggccgagcg gggagacatt ctcctgagca gcctcatcag aaagaagctg 360 cttcccgagg cctctctgct catcaccacg agacctgtgg ccctggagaa actgcagcac 420 ttgctggacc atcctcggca tgtggagatc ctgggtttct ccgaggccaa aaggaaagag 480 tacttcttca agtacttctc tgatgaggcc caagccaggg cagccttcag tctgattcag 540 gagaacgagg tcctcttcac catgtgcttc atccccctgg tctgctggat cgtgtgcact 600 ggactgaaac agcagatgga gagtggcaag agccttgccc agacatccaa gaccaccacc 660 gcggtgtacg tcttcttcct ttccagtttg ctgcagcccc ggggagggag ccaggagcac 720 ggcctctgcg cccacctctg ggggctctgc tctttggctg cagatggaat ctggaaccag 780 aaaatcctgt ttgaggagtc cgacctcagg aatcatggac tgcagaaggc ggatgtgtct 840 gctttcctga ggatgaacct gttccaaaag gaagtggact gcgagaagtt ctacagcttc 900 atccacatga ctttccagga gttctttgcc gccatgtact acctgctg 948 <210> 24 <211> 414 <212> DNA <213> nacht domain of cryopyrin <400> 24 cgccacgttg atgtttttaa actatggctg atgtggaggg caaaggggac taccgggttt 60 gaagcgcatg ttgataaatg tttggagttg gcagagtatt tatacaacat cataaaaaac 120 cgagaaggat atgagatggt gtttgatggg aagcctcagc acacaaatgt ctgcttctgg 180 tacattcctc caagcttgcg tactctggaa gacaatgaag agagaatgag tcgcctctcg 240 aaggtggctc cagtgattaa agccagaatg atggagtatg gaaccacaat ggtcagctac 300 caacccttgg gagacaaggt caatttcttc cgcatggtca tctcaaaccc agcggcaact 360 caccaagaca ttgacttcct gattgaagaa atagaacgcc ttggacaaga ttta 414 <210> 25 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Sense Primer <400> 25 ctcgagtttg tcaaccaaca t 21 <210> 26 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Antisense Primer <400> 26 aagcttttag ttacagtagt tc 22 <210> 27 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Sense Primer <400> 27 ctcgagaact ctgactccga atgc 24 <210> 28 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Antisense Primer <400> 28 aagcttttaa cgcagttccc acca 24 <210> 29 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Sense Primer <400> 29 ctcgagggct ccagcttcct g 21 <210> 30 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Antisense Primer <400> 30 aagctttcac ttgtcggct 19 <210> 31 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Sense Primer <400> 31 ctcgaggcac ctacttcaag t 21 <210> 32 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Antisense Primer <400> 32 aagcttttat caagtcagtg t 21 <210> 33 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> AID Sense Primer <400> 33 ctcgaggaca gcctcttgat gaac 24 <210> 34 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> AID Antisense Primer <400> 34 aagctttcat aacaaaagtc cca 23 <210> 35 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Sense Primer <400> 35 ctcgagcgcc acgttgatgt 20 <210> 36 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Antisense Primer <400> 36 atcgatttat aaatcttgtc c 21 <210> 37 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Sense Primer <400> 37 ctcgaggctc ccctgccgga tac 23 <210> 38 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Antisense Primer <400> 38 aagcttttaa agcccgcggc gct 23 <210> 39 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Sense Primer <400> 39 ctcgagagct cccagattcg t 21 <210> 40 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Antisense Primer <400> 40 aagcttttag tcgtgcttga gagt 24 <210> 41 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Sense Primer <400> 41 ctcgagactc cactcggacc tg 22 <210> 42 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Antisense Primer <400> 42 aagctttcat ggctgtgcaa g 21 <210> 43 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nacht Sense Primer <400> 43 ctcgagactg tggtgttcca g 21 <210> 44 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nacht Antisense Primer <400> 44 aagctttcac agcaggtagt ac 22 <210> 45 <211> 993 <212> DNA <213> Escherichia coli RpoS <400> 45 ttactcgcgg aacagcgctt cgatattcag cccctgcgtt tgcaggattt cgcgcaaacg 60 gcgcaggcct tcaacctgaa tctggcgaac acgttcacgg gtgaggccaa tttcacgacc 120 tacatcttcc agtgttgccg cttcgtaccc cagcaaaccg aatcgacgtg ccagcacttc 180 acgctgtttg gcgttcagct cgaacagcca tttgacgatg ctctgcttca tatcgtcatc 240 ttgcgtggta tcttccggac cgttctcttt ttcatcggcc aggatgtcca gcaacgcttt 300 ttcggaatca ccacccagcg gggtgtctac cgaggtaatg cgctcgttaa gacgaagcat 360 acggctgacg tcatcaactg gcttatccag ttgctctgcg atctcttccg cacttggttc 420 atggtccagc ttatgggaca actcacgtgc ggttcgcagg taaacgttca gctcctttac 480 gatgtgaatc ggcaaacgaa tagtacgggt ttggttcata atcgcccgtt caatcgtctg 540 gcgaatccac caggttgcgt atgttgagaa gcggaaacca cgttccgggt caaacttctc 600 taccgcgcgg atcagcccca ggttgccctc ttcgataagg tccagcaacg ccagaccacg 660 attgccataa cggcgggcaa tttttaccac cagacgcaag ttactctcga tcatccggcg 720 gcgagaggcg acatctccac gcagtgcgcg acgcgcaaaa taaacttctt cttcggccgt 780 taacagtggt gaataaccaa tctcaccaag gtaaagctga gtcgcgtcca acacacgctg 840 tgtggctccc tgcgataaca gttcctcttc ggccaaatcg ttatcactgg gttcctgttc 900 tactaaggcc ttttcgtcaa aaacctcaac tccgttctca tcaaattccg catcttcatt 960 taaatcatga actttcagcg tattctgact cat 993 <210> 46 <211> 180 <212> DNA <213> human minipro-insulin <400> 46 tttgtcaacc aacatttatg tggatcacat ttagtagagg ctttgtatct tgtttgtggt 60 gaacgtggat ttttctatac acctaagaca cgtagatctc ctaatggaaa acgtggtatt 120 gttgaacaat gctgtacatc aatctgttca ttgtatcaac ttgagaacta ctgtaactaa 180 180 <210> 47 <211> 528 <212> DNA <213> human ferritin light chain <400> 47 atgagctccc agattcgtca gaattattcc accgacgtgg aggcagccgt caacagcctg 60 gtcaatttgt acctgcaggc ctcctacacc tacctctctc tgggcttcta tttcgaccgc 120 gatgatgtgg ctctggaagg cgtgagccac ttcttccgcg aattggccga ggagaagcgc 180 gagggctacg agcgtctcct gaagatgcaa aaccagcgtg gcggccgcgc tctcttccag 240 gacatcaaga agccagctga agatgagtgg ggtaaaaccc cagacgccat gaaagctgcc 300 atggccctgg agaaaaagct gaaccaggcc cttttggatc ttcatgccct gggttctgcc 360 cgcacggacc cccatctctg tgacttcctg gagactcact tcctagatga ggaagtgaag 420 ctcatcaaga agatgggtga ccacctgacc aacctccaca ggctgggtgg cccggaggct 480 gggctgggcg agtatctctt cgaaaggctc actctcaagc acgactaa 528 <210> 48 <211> 780 <212> DNA <213> Pseudomonas putida cutinase <400> 48 atggctcccc tgccggatac accgggagcg ccatttccgg ctgtcgccaa tttcgaccgc 60 agtggcccct acaccaccag cagccagagc gaggggccga gctgtcgcat ctatcggccc 120 cgcgacctgg gtcagggggg cgtgcgtcat ccggtgattc tctggggcaa tggcaccggt 180 gccgggccgt ccacctatgc cggcttgcta tcgcactggg caagccacgg tttcgtggtg 240 gcggcggcgg aaacctccaa tgccggtacc gggcgggaaa tgctcgcctg cctggactat 300 ctggtacgtg agaacgacac cccctacggc acctattccg gcaagctcaa taccgggcga 360 gtcggcactt ctgggcattc ccagggtggt ggcggctcga tcatggccgg gcaggatacg 420 agggtgcgta ccacggcgcc gatccagccc tacaccctcg gcctggggca cgacagcgcc 480 tcgcagcggc ggcagcaggg gccgatgttc ctgatgtccg gtggcggtga caccatcgcc 540 tttccctacc tcaacgctca gccggtctac cggcgtgcca atgtgccggt gttctggggc 600 gaacggcgtt acgtcagcca cttcgagccg gtcggtagcg gtggggccta tcgcggcccg 660 agcacggcat ggttccgctt ccagctgatg gatgaccaag acgcccgcgc taccttctac 720 ggcgcgcagt gcagtctgtg caccagcctg ctgtggtcgg tcgagcgccg cgggctttaa 780 780 <210> 49 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT-His6 Sense Primer <400> 49 ctcgaggctc ccctgccgga tac 23 <210> 50 <211> 38 <212> DNA <213> Artificial Sequence <220> <223> CUT-His6 Antisense primer <400> 50 aagcttttag tgatggtgat ggtgatgaag cccgcgct 38 <110> KOREA UNIVERSITY Industry & Academy Collaboration Foundation <120> PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A          FUSION EXPRESSION PARTNER <130> 7P-08-24 <160> 50 <170> KopatentIn 1.71 <210> 1 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> RpoS Sense Primer <400> 1 catatgagtc agaatacgct gaaa 24 <210> 2 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> RpoS Antisense Primer <400> 2 ctcgagctcg cggaacagcg cttc 24 <210> 3 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Sense Primer <400> 3 catatgtttg tcaaccaaca t 21 <210> 4 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Antisense Primer <400> 4 aagcttttag ttacagtagt tc 22 <210> 5 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Sense Primer <400> 5 catatgaact ctgactccga atgc 24 <210> 6 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Antisense Primer <400> 6 aagcttttaa cgcagttccc acca 24 <210> 7 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Sense Primer <400> 7 catatgggct ccagcttcct g 21 <210> 8 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Antisense Primer <400> 8 aagctttcac ttgtcggct 19 <210> 9 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Sense Primer <400> 9 catatggcac ctacttcaag t 21 <210> 10 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Antisense Primer <400> 10 aagcttttat caagtcagtg t 21 <210> 11 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> AID Sense Primer <400> 11 catatggaca gcctcttgat gaac 24 <210> 12 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> AID Antisense Primer <400> 12 aagctttcat aacaaaagtc cca 23 <210> 13 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Sense Primer <400> 13 catatgcgcc acgttgatgt 20 <210> 14 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Antisense Primer <400> 14 atcgatttat aaatcttgtc c 21 <210> 15 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Sense Primer <400> 15 catatggctc ccctgccgga tac 23 <210> 16 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Antisense Primer <400> 16 aagcttttaa agcccgcggc gct 23 <210> 17 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Sense Primer <400> 17 catatgagct cccagattcg t 21 <210> 18 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Antisense Primer <400> 18 aagcttttag tcgtgcttga gagt 24 <210> 19 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Sense Primer <400> 19 catatgactc cactcggacc tg 22 <210> 20 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Antisense Primer <400> 20 aagctttcat ggctgtgcaa g 21 <210> 21 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nacht Sense Primer <400> 21 catatgactg tggtgttcca g 21 <210> 22 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nacht Antisense Primer <400> 22 aagctttcac agcaggtagt ac 22 <210> 23 <211> 948 <212> DNA <213> Human GAD448-585 <400> 23 accgtggtgt tccagggggc ggcagggatt gggaaaacaa tcctggcccg gaagatgatg 60 ttggactggg cgtcggggac actctaccaa gacaggtttg actatctgtt ctatatccac 120 tgtcgggagg tgagccttgt gacacagagg agcctggggg acctgatcat gagctgctgc 180 cccgacccaa acccacccat ccacaagatc gtgagaaaac cctccagaat cctcttcctc 240 atggacggct tcgatgagct gcaaggtgcc tttgacgagc acataggacc gctctgcact 300 gactggcata aggccgagcg gggagacatt ctcctgagca gcctcatcag aaagaagctg 360 cttcccgagg cctctctgct catcaccacg agacctgtgg ccctggagaa actgcagcac 420 ttgctggacc atcctcggca tgtggagatc ctgggtttct ccgaggccaa aaggaaagag 480 tacttcttca agtacttctc tgatgaggcc caagccaggg cagccttcag tctgattcag 540 gagaacgagg tcctcttcac catgtgcttc atccccctgg tctgctggat cgtgtgcact 600 ggactgaaac agcagatgga gagtggcaag agccttgccc agacatccaa gaccaccacc 660 gcggtgtacg tcttcttcct ttccagtttg ctgcagcccc ggggagggag ccaggagcac 720 ggcctctgcg cccacctctg ggggctctgc tctttggctg cagatggaat ctggaaccag 780 aaaatcctgt ttgaggagtc cgacctcagg aatcatggac tgcagaaggc ggatgtgtct 840 gctttcctga ggatgaacct gttccaaaag gaagtggact gcgagaagtt ctacagcttc 900 atccacatga ctttccagga gttctttgcc gccatgtact acctgctg 948 <210> 24 <211> 414 <212> DNA <213> nacht domain of cryopyrin <400> 24 cgccacgttg atgtttttaa actatggctg atgtggaggg caaaggggac taccgggttt 60 gaagcgcatg ttgataaatg tttggagttg gcagagtatt tatacaacat cataaaaaac 120 cgagaaggat atgagatggt gtttgatggg aagcctcagc acacaaatgt ctgcttctgg 180 tacattcctc caagcttgcg tactctggaa gacaatgaag agagaatgag tcgcctctcg 240 aaggtggctc cagtgattaa agccagaatg atggagtatg gaaccacaat ggtcagctac 300 caacccttgg gagacaaggt caatttcttc cgcatggtca tctcaaaccc agcggcaact 360 caccaagaca ttgacttcct gattgaagaa atagaacgcc ttggacaaga ttta 414 <210> 25 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Sense Primer <400> 25 ctcgagtttg tcaaccaaca t 21 <210> 26 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> mp-INS Antisense Primer <400> 26 aagcttttag ttacagtagt tc 22 <210> 27 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Sense Primer <400> 27 ctcgagaact ctgactccga atgc 24 <210> 28 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> EGF Antisense Primer <400> 28 aagcttttaa cgcagttccc acca 24 <210> 29 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Sense Primer <400> 29 ctcgagggct ccagcttcct g 21 <210> 30 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> ppGRN Antisense Primer <400> 30 aagctttcac ttgtcggct 19 <210> 31 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Sense Primer <400> 31 ctcgaggcac ctacttcaag t 21 <210> 32 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hIL-2 Antisense Primer <400> 32 aagcttttat caagtcagtg t 21 <210> 33 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> AID Sense Primer <400> 33 ctcgaggaca gcctcttgat gaac 24 <210> 34 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> AID Antisense Primer <400> 34 aagctttcat aacaaaagtc cca 23 <210> 35 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Sense Primer <400> 35 ctcgagcgcc acgttgatgt 20 <210> 36 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> GAD448-585 Antisense Primer <400> 36 atcgatttat aaatcttgtc c 21 <210> 37 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Sense Primer <400> 37 ctcgaggctc ccctgccgga tac 23 <210> 38 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT Antisense Primer <400> 38 aagcttttaa agcccgcggc gct 23 <210> 39 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Sense Primer <400> 39 ctcgagagct cccagattcg t 21 <210> 40 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> hFTN-L Antisense Primer <400> 40 aagcttttag tcgtgcttga gagt 24 <210> 41 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Sense Primer <400> 41 ctcgagactc cactcggacc tg 22 <210> 42 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> G-CSF Antisense Primer <400> 42 aagctttcat ggctgtgcaa g 21 <210> 43 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Nacht Sense Primer <400> 43 ctcgagactg tggtgttcca g 21 <210> 44 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Nacht Antisense Primer <400> 44 aagctttcac agcaggtagt ac 22 <210> 45 <211> 993 <212> DNA <213> Escherichia coli RpoS <400> 45 ttactcgcgg aacagcgctt cgatattcag cccctgcgtt tgcaggattt cgcgcaaacg 60 gcgcaggcct tcaacctgaa tctggcgaac acgttcacgg gtgaggccaa tttcacgacc 120 tacatcttcc agtgttgccg cttcgtaccc cagcaaaccg aatcgacgtg ccagcacttc 180 acgctgtttg gcgttcagct cgaacagcca tttgacgatg ctctgcttca tatcgtcatc 240 ttgcgtggta tcttccggac cgttctcttt ttcatcggcc aggatgtcca gcaacgcttt 300 ttcggaatca ccacccagcg gggtgtctac cgaggtaatg cgctcgttaa gacgaagcat 360 acggctgacg tcatcaactg gcttatccag ttgctctgcg atctcttccg cacttggttc 420 atggtccagc ttatgggaca actcacgtgc ggttcgcagg taaacgttca gctcctttac 480 gatgtgaatc ggcaaacgaa tagtacgggt ttggttcata atcgcccgtt caatcgtctg 540 gcgaatccac caggttgcgt atgttgagaa gcggaaacca cgttccgggt caaacttctc 600 taccgcgcgg atcagcccca ggttgccctc ttcgataagg tccagcaacg ccagaccacg 660 attgccataa cggcgggcaa tttttaccac cagacgcaag ttactctcga tcatccggcg 720 gcgagaggcg acatctccac gcagtgcgcg acgcgcaaaa taaacttctt cttcggccgt 780 taacagtggt gaataaccaa tctcaccaag gtaaagctga gtcgcgtcca acacacgctg 840 tgtggctccc tgcgataaca gttcctcttc ggccaaatcg ttatcactgg gttcctgttc 900 tactaaggcc ttttcgtcaa aaacctcaac tccgttctca tcaaattccg catcttcatt 960 taaatcatga actttcagcg tattctgact cat 993 <210> 46 <211> 180 <212> DNA <213> human minipro-insulin <400> 46 tttgtcaacc aacatttatg tggatcacat ttagtagagg ctttgtatct tgtttgtggt 60 gaacgtggat ttttctatac acctaagaca cgtagatctc ctaatggaaa acgtggtatt 120 gttgaacaat gctgtacatc aatctgttca ttgtatcaac ttgagaacta ctgtaactaa 180                                                                          180 <210> 47 <211> 528 <212> DNA <213> human ferritin light chain <400> 47 atgagctccc agattcgtca gaattattcc accgacgtgg aggcagccgt caacagcctg 60 gtcaatttgt acctgcaggc ctcctacacc tacctctctc tgggcttcta tttcgaccgc 120 gatgatgtgg ctctggaagg cgtgagccac ttcttccgcg aattggccga ggagaagcgc 180 gagggctacg agcgtctcct gaagatgcaa aaccagcgtg gcggccgcgc tctcttccag 240 gacatcaaga agccagctga agatgagtgg ggtaaaaccc cagacgccat gaaagctgcc 300 atggccctgg agaaaaagct gaaccaggcc cttttggatc ttcatgccct gggttctgcc 360 cgcacggacc cccatctctg tgacttcctg gagactcact tcctagatga ggaagtgaag 420 ctcatcaaga agatgggtga ccacctgacc aacctccaca ggctgggtgg cccggaggct 480 gggctgggcg agtatctctt cgaaaggctc actctcaagc acgactaa 528 <210> 48 <211> 780 <212> DNA <213> Pseudomonas putida cutinase <400> 48 atggctcccc tgccggatac accgggagcg ccatttccgg ctgtcgccaa tttcgaccgc 60 agtggcccct acaccaccag cagccagagc gaggggccga gctgtcgcat ctatcggccc 120 cgcgacctgg gtcagggggg cgtgcgtcat ccggtgattc tctggggcaa tggcaccggt 180 gccgggccgt ccacctatgc cggcttgcta tcgcactggg caagccacgg tttcgtggtg 240 gcggcggcgg aaacctccaa tgccggtacc gggcgggaaa tgctcgcctg cctggactat 300 ctggtacgtg agaacgacac cccctacggc acctattccg gcaagctcaa taccgggcga 360 gtcggcactt ctgggcattc ccagggtggt ggcggctcga tcatggccgg gcaggatacg 420 agggtgcgta ccacggcgcc gatccagccc tacaccctcg gcctggggca cgacagcgcc 480 tcgcagcggc ggcagcaggg gccgatgttc ctgatgtccg gtggcggtga caccatcgcc 540 tttccctacc tcaacgctca gccggtctac cggcgtgcca atgtgccggt gttctggggc 600 gaacggcgtt acgtcagcca cttcgagccg gtcggtagcg gtggggccta tcgcggcccg 660 agcacggcat ggttccgctt ccagctgatg gatgaccaag acgcccgcgc taccttctac 720 ggcgcgcagt gcagtctgtg caccagcctg ctgtggtcgg tcgagcgccg cgggctttaa 780                                                                          780 <210> 49 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> CUT-His6 Sense Primer <400> 49 ctcgaggctc ccctgccgga tac 23 <210> 50 <211> 38 <212> DNA <213> Artificial Sequence <220> <223> CUT-His6 Antisense primer <400> 50 aagcttttag tgatggtgat ggtgatgaag cccgcgct 38  

Claims (9)

1) 융합파트너(fusion partner)로서 RpoS(RNA polymerase sigma factor)의 유전자 및 외래단백질의 유전자를 연결한 발현벡터를 제조하는 단계;1) preparing an expression vector linking a gene of RpoS (RNA polymerase sigma factor) and a gene of a foreign protein as a fusion partner; 2) 상기 발현벡터를 숙주세포에 도입하여 형질전환체를 제조하는 단계; 및,2) preparing a transformant by introducing the expression vector into a host cell; And, 3) 상기 형질전환체를 배양하여 재조합 단백질의 발현을 유도하고 이를 수득하는 단계로 구성되는 재조합 단백질의 제조방법.3) culturing the transformant to induce the expression of the recombinant protein and the method of producing a recombinant protein consisting of obtaining it. 제 1항에 있어서, RpoS의 유전자는 서열번호 45로 기재되는 것을 특징으로 하는 재조합 단백질의 제조방법.The method of claim 1, wherein the gene of RpoS is set forth in SEQ ID NO: 45. 제 1항에 있어서, 외래단백질은 항원, 항체, 세포수용체, 효소, 구조 단백질, 혈청 및 세포 단백질로 이루어진 군으로부터 선택되는 생물학적 활성을 갖는 것을 특징으로 하는 재조합 단백질의 제조방법.The method of claim 1, wherein the foreign protein has a biological activity selected from the group consisting of antigens, antibodies, cell receptors, enzymes, structural proteins, serum and cellular proteins. 융합파트너로서 RpoS의 유전자와 외래단백질의 유전자를 포함하는 재조합 단백질 생산용 발현벡터.An expression vector for recombinant protein production comprising a gene of RpoS and a gene of foreign protein as a fusion partner. 제 4항에 있어서, 도 2의 개열지도로 표시되는 것을 특징으로 하는 발현벡터.The expression vector according to claim 4, which is represented by the cleavage map of FIG. 제 4항에 있어서, 융합파트너로서 RpoS 유전자와 외래단백질의 유전자가 연결되어 포함되는 것을 특징으로 하는 발현벡터.The expression vector according to claim 4, wherein the fusion partner includes an RpoS gene and a foreign protein gene. 제 4항에 있어서, RpoS 유전자와 외래단백질 유전자 사이에 단백질 절단효소 인식부위를 코딩하는 폴리뉴클레오티드가 연결되는 것을 특징으로 하는 발현벡터.The expression vector according to claim 4, wherein a polynucleotide encoding a protein cleavage site is linked between the RpoS gene and the foreign protein gene. 제 4항의 발현벡터가 형질 도입된 형질전환체.The transformant transformed with the expression vector of claim 4. 1 항의 방법에 의해 제조된 재조합 융합 단백질.A recombinant fusion protein produced by the method of claim 1.
KR1020070090368A 2007-09-06 2007-09-06 PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER KR100890186B1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
KR1020070090368A KR100890186B1 (en) 2007-09-06 2007-09-06 PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER
PCT/KR2008/005256 WO2009031852A2 (en) 2007-09-06 2008-09-05 Preparation method of recombinant protein by use of a fusion expression partner
US12/676,982 US20100210827A1 (en) 2007-09-06 2008-09-05 Preparation Method Of Recombinant Protein By Use Of A Fusion Expression Partner
US13/108,518 US20110281300A1 (en) 2007-09-06 2011-05-16 Preparation method of recombinant protein by use of a fusion expression partner

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020070090368A KR100890186B1 (en) 2007-09-06 2007-09-06 PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER

Publications (2)

Publication Number Publication Date
KR20090025481A true KR20090025481A (en) 2009-03-11
KR100890186B1 KR100890186B1 (en) 2009-03-25

Family

ID=40693808

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020070090368A KR100890186B1 (en) 2007-09-06 2007-09-06 PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER

Country Status (1)

Country Link
KR (1) KR100890186B1 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5670350A (en) 1990-08-20 1997-09-23 Novartis Finance Corporation Genomic DNA encoding a pseudomonas global transcriptional activation element and its use in activating gene expression

Also Published As

Publication number Publication date
KR100890186B1 (en) 2009-03-25

Similar Documents

Publication Publication Date Title
EP1924601B1 (en) Expression of proteins in e.coli
IE63991B1 (en) Novel fusion proteins and their purification
CN103952425A (en) Screening of abundantly secreted proteins and their use as fusion partners for the production of recombinant proteins
KR101373297B1 (en) Expression Vector Comprising Gene coding for E. coli Phosphoglycerate kinase As a Novel Fusion Partner
KR100890184B1 (en) PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF SlyD AS A FUSION EXPRESSION PARTNER
KR100890187B1 (en) PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF Tsf AS A FUSION EXPRESSION PARTNER
KR100890183B1 (en) Preparation method of recombinant protein by use of malate dehydrogenase as a fusion expression partner
KR100890186B1 (en) PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF RpoS AS A FUSION EXPRESSION PARTNER
KR100890188B1 (en) PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF PotD AS A FUSION EXPRESSION PARTNER
KR100890185B1 (en) PREPARATION METHOD OF RECOMBINANT PROTEIN BY USE OF Crr AS A FUSION EXPRESSION PARTNER
KR100890189B1 (en) Preparation method of recombinant protein by use of rna polymerase ?subunit as a fusion expression partner
KR101658081B1 (en) Method for Preparing Recombinant Protein Using CysQ as a Fusion Expression Partner
EP2643457B1 (en) Compositions and methods of producing enterokinase in yeast
KR101077611B1 (en) A preparation method of solubility recombinant protein by use of Chemotaxis protein cheZ as a fusion expression partner
KR101658082B1 (en) Method for Preparing Recombinant Protein Using EDA as a Fusion Expression Partner
KR101077612B1 (en) A preparation method of solubility recombinant protein by use of Bacterioferritin as a fusion expression partner
US20110281300A1 (en) Preparation method of recombinant protein by use of a fusion expression partner
KR20160088259A (en) Method for Preparing Active Arginine Deiminase
EP4079845A1 (en) Method for enhancing water solubility of target protein by whep domain fusion
KR101023518B1 (en) A preparation method of solubility recombinant protein by use of Aspartate carbamoyltransferase catalytic chain as a fusion expression partner
KR20120006002A (en) A preparation method of solubility recombinant protein by use of dihydrofolate reductase as a fusion expression partner
JPH0928380A (en) Control factor related to expression of nitrilase gane and the gane
KR20100077665A (en) A preparation method of solubility recombinant protein by use of peptidyl-prolyl cis-trans isomerase b as a fusion expression partner
KR20160088014A (en) Method for Preparing Active Arginine Deiminase
KR20240072296A (en) Extracellular secretion system of recombinant protein using monomeric DsRed and intein variant

Legal Events

Date Code Title Description
A201 Request for examination
E701 Decision to grant or registration of patent right
GRNT Written decision to grant
FPAY Annual fee payment

Payment date: 20130311

Year of fee payment: 5

FPAY Annual fee payment

Payment date: 20140312

Year of fee payment: 6

FPAY Annual fee payment

Payment date: 20150315

Year of fee payment: 7

FPAY Annual fee payment

Payment date: 20170405

Year of fee payment: 9

LAPS Lapse due to unpaid annual fee