WO2014056199A1 - 多肽的产生和纯化方法 - Google Patents

多肽的产生和纯化方法 Download PDF

Info

Publication number
WO2014056199A1
WO2014056199A1 PCT/CN2012/082879 CN2012082879W WO2014056199A1 WO 2014056199 A1 WO2014056199 A1 WO 2014056199A1 CN 2012082879 W CN2012082879 W CN 2012082879W WO 2014056199 A1 WO2014056199 A1 WO 2014056199A1
Authority
WO
WIPO (PCT)
Prior art keywords
fusion protein
peptide
self
polypeptide
amino acid
Prior art date
Application number
PCT/CN2012/082879
Other languages
English (en)
French (fr)
Inventor
林章凛
徐望晖
邢磊
曾伶俐
周碧红
赵青
吴伟
Original Assignee
清华大学
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 清华大学 filed Critical 清华大学
Priority to PCT/CN2012/082879 priority Critical patent/WO2014056199A1/zh
Priority to CN201280076298.3A priority patent/CN104755502B/zh
Priority to US13/836,817 priority patent/US9200306B2/en
Publication of WO2014056199A1 publication Critical patent/WO2014056199A1/zh

Links

Classifications

    • 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
    • C12P21/06Preparation of peptides or proteins produced by the hydrolysis of a peptide bond, e.g. hydrolysate products
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • 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
    • C07K14/575Hormones
    • C07K14/57581Thymosin; Related peptides
    • 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
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • 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
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction

Definitions

  • the invention relates to the field of genetic engineering. More specifically, the present invention relates to a fusion protein comprising a pro-lysed peptide tag moiety, a self-aggregating peptide moiety and a polypeptide moiety of interest, and a method of producing and purifying a polypeptide of interest by expressing the fusion protein.
  • a fusion protein comprising a pro-lysed peptide tag moiety, a self-aggregating peptide moiety and a polypeptide moiety of interest, and a method of producing and purifying a polypeptide of interest by expressing the fusion protein.
  • Another effective means is the use of recombinant methods to produce polysaccharides in host cells.
  • Escherichia coli With high growth rate, high expression amount and low production cost, it is easy to produce the desired multi-purine product by introducing foreign genes or operating the amino acid sequence, and the method is simple and easy to operate. Zoom in production. More than 30% of recombinant peptides currently on the market are produced using E. coli cells (Kamionka et al, 2011; Demain et al, 2009; Walsh, 2003 and 2006).
  • the purification step is critical. It has been reported that the cost of separation and purification of recombination is about 60% to 80% of its total production cost (Chen Hao et al., 2002).
  • Methods for purifying recombinant polypeptides include conventional ion exchange chromatography, hydrophobic interaction chromatography, affinity chromatography, and the like. Ion exchange chromatography and hydrophobic interaction chromatography are less versatile and inefficient than affinity chromatography due to certain requirements for the initiation conditions of the sample. Affinity purification typically yields high yields of over 90%, making it the most commonly used recombinant protein purification method.
  • affinity purification techniques include the fusion expression of a histidine tag or a glutathione transferase tag (GSTTtag) with a polypeptide of interest, providing a universal purification means for the production of polysaccharides of different purposes.
  • GSTTtag glutathione transferase tag
  • the present invention provides a low cost, simple, and efficient multi-aggregation peptide and solubilization-based label.
  • the present invention provides a fusion protein comprising a solubilizing moiety, a self-aggregating peptide moiety, and a polypeptide moiety of interest, wherein the polypeptide moiety of interest is located in the lytic peptide tag moiety and the self Between the peptide moieties, and the polypeptide moiety of interest is linked to the self-aggregating peptide moiety by a first linker, wherein the first linker comprises a first cleavage site, and the fusion protein is expressed in a host cell
  • the self-aggregating peptide moiety forms an active aggregate.
  • the self-aggregating peptide moiety is located at the C-terminus of the fusion protein.
  • the self-aggregating peptide moiety comprises an amphipathic self-assembling short peptide.
  • the amphiphilic self-assembling short peptide is selected from the group consisting of an amphiphilic beta-sheet short peptide, an amphipathic alpha-helical short peptide, and a surfactant-like short peptide.
  • the amphiphilic beta-sheeted short peptide is 4-30 amino acid residues in length. In some embodiments, the amphiphilic beta-sheeted short peptide has a hydrophobic amino acid residue content of from 40% to 80%. In a specific embodiment, the amino acid sequence of the amphiphilic beta-sheeted short peptide is shown in SEQ ID NO. In some embodiments, the self-aggregating peptide portion of the fusion protein of the invention comprises one of the amphipathic beta Fold the short peptide. In other embodiments, the self-aggregating peptide portion of the fusion protein of the invention comprises two or more of said amphiphilic beta-sheeted short peptides in tandem repeats.
  • the amphipathic alpha helix short peptide is 4-30 amino acid residues in length. In some embodiments, the amphiphilic alpha helix short peptide has a hydrophobic amino acid residue content of from 40% to 80%. In a specific embodiment, the amino acid sequence of the amphipathic alpha helix short peptide is shown in SEQ ID NO. In some embodiments, the self-aggregating peptide portion of the fusion protein of the invention comprises one of the amphipathic alpha Spiral short peptide. In other embodiments, the self-aggregating peptide portion of the fusion protein of the invention comprises two or more of said amphipathic alpha helical short peptides in tandem.
  • the surfactant-like short peptide has 7-30 amino acid residues having an amino acid sequence represented by the following formula from the quinone to the C-terminus: AB or BA
  • AA is a peptide peptide composed of a hydrophilic hydrophilic aminoamino acid acid residue group, and the hydrophilic/hydrophilic aminoamino acid residue may be Is identical or not identical, and is selected from the group consisting of LMyyss, AAsspp.. AArrgg, GGlluu, HHiiss.. SSeerr, TThhrr, AAssnn, and GGiinn;
  • BB is a peptide peptide composed of a hydrophobic aqueous amino amino acid residue group, and the hydrophobic amino acid amino acid residue may be a phase The same same 55 or different, and selected from LLeeuu, GGllyy, AAllaa.. VVaall, IIllee, PPhhee and TTiipp;; and AA and BB pass a peptide peptide bond linkage; and and wherein a hydrophobic amino acid amino acid residue is present in the active peptide short peptide peptide on the surface of the class surface
  • the ratio of the ratio is 5555%% - 9955%%. .
  • the surface active surfactant short peptide peptide has 88 aminoamino acid residues.
  • the specific proportion of the hydrophobic aqueous amino amino acid residue in the middle is 7755% by weight.
  • the sequence of the amino amino acid sequence of the surface active surfactant short peptide peptide is shown. For SSEEQQ IIDD NNOO::33, SSEEQQ IIDD NNOO::44 or 1100 SSEEQQ IIDD NNOO::55. .
  • the lysopeptide peptide labeling in the fusion fusion egg white of the present invention is selected from the group consisting of NnuussAA, GGSSTT, and TTrrxx. , SSUUMMOO,, DDssbbCC, ZZ, GGBB11, MMBBPP and TT77PPKK. .
  • the first all-cleavage cleavage site selected from the fusion fusion protein protein of the present invention is selected from the group consisting of Chemical chemistry cuts the cutting site, enzymatically cuts the cleavage site, and self-cuts the cleavage site. .
  • the self-cutting 1155 cutting site is intron-containing iniinnteteiinn)).
  • the endo-containing peptide-containing peptide is MMxxee GGyyrrAA shown in the sequence of SSEEQQ IIDD NNOO::66. .
  • MMxxee GGyyrrAA is ligated directly to the CC terminus of the polypeptide peptide of the stated purpose. .
  • the first first joint head of the fusion fusion protein protein of the present invention is further The package contains interstitial spacers. .
  • the fused fusion egg protein white of the present invention has been passed through the second and second The linker is ligated to the lytic peptide label tag as described.
  • the second and second joint head packs comprise interstitial spacers.
  • the second second joint head package includes a second second cutting cut position point, The cutting cutting condition condition of the second second cutting cutting position point is not the condition of the cutting cutting cutting condition of the first all cutting cutting position point.
  • the second second-cutting cut site is selected from the second second chemical cleavage site, The second two enzyme enzymatic method
  • the second enzyme enzymatically cutting the cleavage site contains a SSEEQQ IIDD NNOO::77
  • the indicated enterokinase kinase recognizes the sequence of the sequence.
  • the enterokinase kinase recognition sequence is directly connected in series to the stated statement.
  • the second and second joint heads are packaged in addition to the second second cutting position. Includes intervening spacers. .
  • the multi-peptide peptide of the above-mentioned purpose of the fusion fusion protein protein of the present invention is characterized by a long length of
  • the present invention provides a multi-nuclear nucleoside acid, which comprises a coding code comprising the above-described invention.
  • the invention provides an expression construct comprising the polynucleotide of the invention described above.
  • the invention provides a host cell comprising or transformed with a polynucleotide of the invention described above, wherein said host cell is capable of expressing a fusion protein of the invention described above.
  • the invention provides a method of producing and purifying a polypeptide of interest, the method comprising the steps of:
  • the method of producing and purifying a polypeptide of interest of the invention further comprises:
  • FIG. 1 Self-aggregating peptide (ELK16) and lytic peptide labeling ( ⁇ multi-purine production and purification strategy and expression vector map.
  • band-promoting peptide tag
  • without lytic peptide tag
  • D pET Trx-EK- lutein-ELK16 vector.
  • FIG. 1 Expression and purification results of the polypeptide of interest. A: Without Trx label; with Trx label.
  • Figure 6. Results of the combination of the lysin-loaded peptide GST and the self-aggregating peptide ELK16 for the production and purification of the desired polysaccharide.
  • Figure 7. Results of the combination of the solubilized J3 ⁇ 4 tag DsbC in combination with the self-aggregating peptide ELK16 for the production and purification of polyhydrazine.
  • Figure 8 Stimulating hydrazine label The results of GB1 in combination with the self-aggregating peptide ELK16 for the production and purification of peptides of interest.
  • the present invention provides a fusion protein comprising a solubilizing oxime tag moiety, a self-aggregating peptide moiety, and a polypeptide moiety of interest, wherein the polypeptide moiety of interest is located in the lytic peptide tag moiety and the self-aggregating Between the peptide moieties, and the polypeptide moiety of interest is linked to the self-aggregating peptide moiety by a first linker, wherein the first linker comprises a first cleavage site, and the fusion protein can be expressed in a host cell
  • the self-aggregating peptide moiety forms an active aggregate.
  • the invention provides a polynucleotide comprising a nucleotide sequence encoding a fusion protein of the invention or a complement thereof.
  • the invention provides an expression construct comprising a polynucleotide of the invention.
  • the invention provides a host cell comprising a polynucleotide of the invention or transformed with an expression construct of the invention, wherein said host cell is capable of expressing a fusion protein of the invention.
  • the invention provides a method of producing and purifying a polypeptide of interest, the method comprising the steps of: (a) cultivating a host cell of the invention to express the fusion protein; (b) lysing the host cell And then removing the soluble portion of the cell lysate, recovering the insoluble portion; (c) releasing the soluble polypeptide having the lytic peptide tag from the insoluble portion by cleaving the first cleavage site; and (d) removing the step The insoluble portion in (C) recovers a soluble portion containing the polypeptide of interest.
  • polysaccharide or “protein of interest” refers to any polypeptide or protein that can be produced and purified by the methods of the invention, non-limiting examples of which include enzymes, hormones, immunoglobulin chains, such as anti-cancer polypeptides. It is more therapeutic) 3 ⁇ 4, more diagnostic) 3 ⁇ 4 or can be used to immunize more than 3 ⁇ 4 or its biologically active fragments and so on.
  • the polypeptide may be from any source, including microbial-derived polypeptides, merinoside-derived polypeptides, and artificial proteins (such as fusion proteins or mutated proteins) and the like.
  • the polypeptide of interest can be a polypeptide and protein of any length.
  • the polypeptide of interest which can be produced and purified by the methods of the invention can be 20-200, 25-150, 30-120, 30-100 amino acid residues, for example, about 30, about 40, about 50, about 60, Approximately 70, approximately 80, approximately 90 amino acid residues.
  • target polysaccharides which can be produced and purified by the method of the present invention include, but are not limited to, glucagon-like peptide GLP-i, B-type natriuretic peptide BNP, prostaglandin secreting peptide Ex-4, Chemokine CCL5, stromal cell-derived factor SDF-1 ⁇ , growth-promoting factor IGF-ia, obesity hormone Lep, calcitonin, 'Sermordin', 'chest, gland H (Thymosin) ), X-ephedrine (Lepimdin), Cecoropin, human rich histone (Histatin), defensin (defensin), and human plasminogen (Kringle i-5) or biologically active fragments thereof Wait.
  • Chemokine CCL5 stromal cell-derived factor SDF-1 ⁇
  • growth-promoting factor IGF-ia obesity hormone Lep, calcitonin, 'Sermordin', 'chest, gland H (Th
  • pro-lysed peptide tag refers to a fusion tag that, when fused to a polypeptide of interest, can help the target to fold properly and increase the solubility of the fusion protein.
  • Many such "pro-lysed peptide tags” are known to those skilled in the art.
  • Suitable solubilizing peptide tags useful in the present invention include, but are not limited to,
  • the solubilizing peptide tag is Trx.
  • self-aggregating peptide refers to a polypeptide that is fused to a polypeptide of interest and that, upon expression in a host cell, is capable of mediating the formation of an insoluble active aggregate in the cell of the fusion protein.
  • Active aggregate means that the portion of the polypeptide of interest is still correctly folded and remains active or that the portion of the polypeptide of interest in the aggregate is capable of being soluble after separation from the self-aggregating mites.
  • amphipathic polypeptides can spontaneously form specific ones due to hydrophobic interactions and other motivating forces due to hydrophilic and hydrophobic regions that are separated from one another.
  • Self-assembled structure Zhang et al., 2008.
  • the inventors have surprisingly discovered that some amphiphilic short peptides having self-assembly ability are capable of inducing the formation of active aggregates in cells.
  • amphiphilic self-assembling short peptide used as the self-aggregating peptide of the present invention may be selected from the group consisting of an amphiphilic ⁇ -sheet short peptide, an amphiphilic ⁇ -helical short peptide, and a surfactant-like short peptide.
  • amphiphilic ⁇ -sheet short peptide refers to a short peptide consisting of 4 to 30 amino acid residues alternately arranged by a hydrophobic amino acid or a charged hydrophilic amino acid, which forms a ⁇ -sheet, The side is a hydrophobic amino acid residue, and the other side is an alternately arranged positively charged and negatively charged pro Aqueous amino acid residues.
  • These short peptides can form self-assembled structures under hydrophobic interactions, electrostatic interactions, and hydrogen bonding. In general, the longer the length of the amphiphilic ⁇ -sheet structure or the stronger the hydrophobicity, the easier the self-assembly occurs, and the stronger the mechanical strength of the formed self-aggregates.
  • amphiphilic ⁇ -sheet short peptide of the present invention should contain a certain amount of hydrophobic amino acid.
  • the amphiphilic beta-sheet short peptide of the present invention comprises 40-80%, 45-70%, 50-60%, for example about 50% of hydrophobic amino acid residues.
  • a specific example of the amphiphilic ⁇ -sheet short peptide which can be used as the self-aggregating peptide of the present invention is ELK16 (amino acid sequence: LELELKLKLELELKLK) having the amino acid sequence shown in SEQ ID NO: 1.
  • a polypeptide having self-aggregating properties is formed by tandem repeating of multiple repeating units, such as elastin-like ELP, which consists of 110 VPGXG repeating units whose aggregation properties are related to the number of repeating units (Banki, et al. , 2005; MacEwan and Chilkoti, 2010). It has also been reported that the self-aggregation tendency of amphipathic ⁇ -sheets composed of multiple repeating units increases as the number of repeat units increases (Zhang et al, 1992). It is expected that a polypeptide consisting of a plurality of the above-mentioned "amphiphilic ⁇ -folded short peptides" in series can retain or even obtain enhanced self-assembly ability.
  • the self-aggregating peptide portion of the invention may comprise one or more of the amphiphilic beta-sheeted short peptides linked in series.
  • the self-aggregating peptide portion of the present invention may comprise 1-150, 1-130, 1-110, 1-90, 1-70, 1-50, 1-30, 1-10, 1-5, for example 1, 2, 3, 4, 5 of the amphipathic ⁇ -sheet short peptides.
  • Two or more amphiphilic ⁇ -sheet short peptides in the self-aggregating peptide moiety may form a tandem repeat. In order to facilitate reorganization operations and to take into account production cost issues, it is desirable to use less repetition.
  • the "self-aggregating peptide moiety" comprises only one of the amphiphilic beta-sheeted short peptides.
  • the alpha helix is a protein secondary structure in which the peptide chain backbone extends in a spiral around an axis.
  • amphiphilic alpha helix short peptide means having 4 to 30 amino acid residues, having a unique hydrophilic, hydrophobic amino acid arrangement compared to a conventional alpha helix, such that one side of the alpha helix structure is formed.
  • a short peptide mainly composed of a hydrophilic amino acid and a hydrophobic amino acid on the other side.
  • amphipathic alpha helix achieves self-assembly by forming a coiled-coil in aqueous solution, wherein the two alpha helices are bound by hydrophobic interactions and further stabilize this binding by the electrostatic interaction forces of the charged amino acids.
  • the amphiphilic alpha helix short peptide of the present invention comprises 40-80%, 45-70%, 50-60%, for example about 50% of hydrophobic amino acid residues.
  • a specific example of the amphiphilic ⁇ -helical short peptide which can be used as the self-aggregating peptide of the present invention is the amino acid sequence shown in 18 A of SEQ ID NO: 2 (EWLKAFYEKVLEKLKELF).
  • a polypeptide consisting of an amphipathic alpha helix short peptide in series maintains or even enhances self-assembly ability.
  • the "self-aggregating peptide moiety" of the present invention may comprise one or more of the amphipathic alpha helix short peptides linked in series.
  • the "self-aggregating peptide moiety” of the present invention may comprise 1-150, 1-130, 1-110, 1-90, 1-70, 1-50, 1-30, 1-10, 1-5
  • 1, 2, 3, 4, and 5 of the amphipathic ⁇ -helical short peptides For example, 1, 2, 3, 4, and 5 of the amphipathic ⁇ -helical short peptides.
  • a "self-aggregating peptide moiety" of the invention comprises only one of the amphipathic alpha helix short peptides.
  • a "surfactant-like peptide” is another type of amphiphilic polypeptide which can be used as the self-aggregating peptide of the present invention, which usually consists of 7-30 amino acid residues, has an elongation length of about 2-5 nm, and has a structure similar to a lipid. , consists of a hydrophobic amino acid tail and a hydrophilic amino acid head.
  • the structure of the surfactant-like structure is similar to that of a surfactant, and an assembly structure such as micelles, nanotubes, or the like can be formed in an aqueous solution.
  • the surfactant-like short peptide suitable for use as the self-aggregating peptide of the present invention may be 7 to 30 amino acid residues including an amino acid sequence represented by the following formula from the N-terminus to the C-terminus:
  • a and B are linked by a peptide bond.
  • A is a hydrophilic head composed of a hydrophilic amino acid, and the hydrophilic amino acid residues may be the same or different and are selected from the group consisting of Lys, Asp, Arg, Glu, His. Ser, Thr, Asn, and Gln. Examples of A include KD, KK, and the like.
  • B is a hydrophobic tail composed of hydrophobic amino acid residues, which may be the same or different and selected from the group consisting of Leu, Gly, Ala, Val, Ile, Phe and Trp. Examples of B include LLLLLL (L6) or GAVIL and the like.
  • the ratio of the hydrophobic amino acid in the surfactant-like short peptide of the present invention is higher than the ratio of the hydrophilic amino acid, and the ratio of the hydrophobic amino acid in the surfactant-like short peptide may be 55-95%, 60-95%, 65-95%, 70-95%, 75-95%, 80-95%, 85-95%, 90-95%.
  • the surfactant-like short peptide has 8 amino acid residues, wherein the ratio of hydrophobic amino acids is 75%.
  • surfactant-like short peptide suitable for the self-aggregating peptide of the present invention include L6KD, L6K2 or DKL6 whose amino acid sequences are shown in SEQ ID NO: 3, SEQ ID NO: 4 or SEQ ID NO: 5, respectively.
  • protein domains such as beta amyloid peptide, VP1, MalE31, CBD d . s, etc.
  • beta amyloid peptide can also induce fusion proteins to form aggregates, and it is contemplated by the present invention that such domains can also be used as "self-aggregating peptides" of the present invention.
  • the structure of these domains is relatively complex and the mechanism by which they induce aggregation is unclear (Mitraki, 2010). It is preferred in the present invention to use a relatively simple structure and Short amphiphilic self-assembling short peptides.
  • a self-aggregating peptide having the ability to induce the formation of active aggregates can form an insoluble protein.
  • active aggregates such as an amphiphilic self-assembling peptide and a polypeptide of interest as a fusion protein expressed in a host cell
  • the formation of aggregates can prevent the degradation of the fusion protein by intracellular proteases, thus increasing the yield of the desired polysaccharides.
  • insoluble aggregates can be collected from cell lysates simply by centrifugation or filtration. Purification of the fusion protein to obtain a preliminary purification of the fusion protein.
  • the soluble fraction containing the polypeptide of interest is insoluble.
  • Part (precipitation) is released, distributed in the supernatant, and the insoluble impurities can be removed by simple centrifugation or filtration, and the soluble polypeptide of interest can be harvested.
  • the polypeptide is produced by such a method based on self-aggregation.
  • the schematic diagram is shown in Figure 1A). The separation and purification steps can be carried out in a tube, avoiding the use of Purified by column significantly reduce production costs.
  • lytic peptide tags can increase the solubility of fusion proteins, as opposed to the tendency of self-aggregating ⁇ to render the fusion protein insoluble.
  • the inventors have surprisingly found that when the fusion protein comprising the lytic peptide tag moiety, the polypeptide moiety of interest and the self-aggregating purine moiety can form insoluble aggregates after expression in the host cell, it is still easy to pass the above steps. Isolation and purification (schematically shown in Figure 1B), while the solubilized peptide tag can further promote the correct folding of the target polypeptone, and can also maintain or enhance the solubility of the polypeptide of interest after removal of the self-aggregating peptide moiety.
  • lytic peptide label and self-aggregating enthalpy can form a functional balance.
  • solubilized peptide tag can also improve the expression of a fusion protein and can be used to produce certain polypeptides which were previously difficult to recombinantly produce.
  • the polypeptide of interest is linked to the self-aggregating purine moiety by a first linker, wherein the first linker comprises a first cleavage site.
  • a "cleavage site” includes sequences required to effect cleavage, such as a proteoglycan recognition sequence for enzymatic cleavage, an intron sequence for self-cleavage, and the like.
  • the first cleavage site of the present invention for releasing a soluble fraction comprising a polypeptide of interest from an insoluble fraction (precipitate) comprises a cleavage site which can be chemically cleaved, enzymatically cleaved or self-cleaved, or a person skilled in the art Any other cleavage site known.
  • a preferred first cleavage site in the present invention can be self-cleavable, for example, comprising an amino acid sequence of a self-cleavable intein.
  • the intein-based cleavage method does not require the addition of an enzyme or the use of harmful substances such as hydrogen bromide used in chemical methods, but only needs to change the buffer environment in which the aggregate is located to induce the cutting alone.
  • a variety of self-cleaving introns are known in the art, such as a series of inteins with different self-cleavage properties from NEB Corporation.
  • the intein is Mxe GyrA having the sequence shown in SEQ ID NO: 6, and the intein can be induced in its carboxyl group by adding an appropriate amount of dithiothreitol to the buffer system.
  • the self-cleavage of the protein of interest in the fusion protein of the present invention may be directly linked to the solubilized peptide tag or may be linked to the solubilizing agent tag via a second linker. If it is desired to obtain a polypeptide of interest that is not labeled, a second cleavage site can be introduced into the second linker. After performing the first cleavage step to separate the target polypeptone from the self-aggregating peptide moiety, the cleavage of the target polypeptidium and the solubilized peptide tag is further separated by cleavage of the second cleavage site, and further purified by further separation (eg, by HPLC). , obtaining a polypeptide of interest without labeling.
  • the method for producing and purifying the present invention may further comprise: (d) if a second cleavage site is present, separating the polypeptide of interest and the lytic peptide label by cleavage of the second cleavage site; (e) removing The solubilizing oxime label is obtained for purification purposes.
  • the second cleavable site of the invention for isolating the target polypeptidic and lytic peptide tags comprises a cleavage site that can be chemically cleaved, enzymatically cleaved or self-cleaved, or any other cleavage site known to those skilled in the art. .
  • the second cleavage site is an enzymatic cleavage site.
  • the enzymatic cleavage site comprises an enterokinase recognition site (amino acid sequence: DDDDK, SEQ ID NO: 7) o
  • the second cleavage site should have a different cutting condition than the first cleavage site such that the second cleavage site is not cleaved when the first cleavage site is cleaved, and the target protein remains associated with the solubilized peptide tag. Connected.
  • This can be achieved in a number of ways, including, for example, the first cleavage site is self-cleaving and the second cleavage site is enzymatically cleaved, or both the first and second cleavage sites are enzymatically cleaved But by different enzymes to cut and so on.
  • spacer refers to a polypeptide having a length consisting of a low hydrophobicity and a low charge effect amino acid, which is used in a fusion protein to allow the joined portions to fully expand and fully fold into each other without interference. Their respective natural conformations.
  • the first linker and/or the second linker may additionally comprise a spacer in addition to the cleavage site.
  • the second linker in the fusion protein of the invention may comprise only spacers and no cleavage sites, as without the need to remove the lytic peptide tag.
  • Such spacers commonly used in the art include, for example, the flexibility of glycine (G) and serine (S).
  • GS-type spacer a rigid PT-type spacer rich in proline (P) and threonine 0. Since the PT type spacer generally has better peptone resistance with respect to the GS type spacer, it is preferred to use the PT type spacer in the present invention.
  • the spacer used in the invention is a PT-type spacer of the order PTPPTTPTPPTTPTPT (SEQ ID NO: 8).
  • the recombinantly produced polypeptide has a sequence identical to the polypeptide of interest, i.e., there are no additional amino acid residues at both ends.
  • this can be achieved by selecting suitable first and second cleavage sites and their attachment to the polypeptide. Those skilled in the art will know how to make such a choice based on the characteristics of the cleavage site.
  • the Mxe GyrA of the first cleavage site can be directly ligated to the C-terminus of the desired ruthenium such that there is no additional amino acid residue between it and the target ruthenium.
  • the cleaved target polypeptide has no extra amino acid residues at the C-terminus.
  • the enterokinase recognition site of the second cleavage site can be directly ligated to the N-terminus of the desired ruthenium such that there are no additional amino acid residues between the polypeptide and the polypeptide of interest. Since enterokinase cleaves directly at the C-terminus of its recognition site, the finally obtained polypeptide of interest has no excess amino acid residues at the N-terminus.
  • the Mxe GyrA of the first cleavage site can be directly linked to the C-terminus of the target polypeptidase and the second cleavage site can be made
  • the enterokinase recognizes that the site is directly linked to the terminus of the polypeptide of interest.
  • the present invention also relates to a polynucleotide comprising a nucleotide sequence encoding the fusion protein of the present invention or a complement thereof.
  • polynucleotide refers to a macromolecule of a plurality of nucleotides joined by a 3'-5'-diacid diester bond, wherein the nucleotide comprises a ribonucleotide and a deoxyribose nucleus. Glycosylate.
  • the sequence of the polynucleotide of the present invention can be codon optimized for different host cells (e.g., E. coli) to improve expression of the fusion protein. Methods for performing codon optimization are known in the art.
  • the present invention also relates to an expression construct comprising the above-described polynuclear acid of the present invention.
  • the sequence of the polynucleotide encoding the fusion protein is operably linked to an expression control sequence for the desired transcription and ultimately production of the fusion protein in a host cell.
  • Suitable expression control sequences include, but are not limited to, promoters, enhancers, ribosome sites of action such as ribosome binding sites, polyadenylation sites, transcriptional splicing sequences, transcription termination sequences, and sequences that stabilize mRNA, and the like.
  • Vectors for use in the expression constructs of the invention include those that replicate autonomously in a host cell.
  • a vector such as a plasmid vector; also includes a vector that is capable of integrating into the host cell DNA and replicating with the host cell DNA.
  • Many vectors suitable for the present invention are commercially available.
  • the expression constructs of the invention are derived from pET30a (+) from Novagen.
  • the invention also relates to a host cell comprising or transformed with a polynucleotide of the invention, wherein said host cell is capable of expressing a fusion protein of the invention.
  • Host cells for expressing the fusion proteins of the invention include prokaryotes, yeast, and higher eukaryotes.
  • Exemplary prokaryotic hosts include the genus Escherichia (c/ien'di i, Bacillus ( ⁇ «7 ⁇ , Salmonella (5 ⁇ wi. "e ⁇ a), and Pseudomonas ( ⁇ £? ⁇ [.
  • the host cell used was E. coli BL21 (DE3) strain cells (Novagen).
  • the recombinant expression constructs of the invention can be introduced into a host cell by one of many well-known techniques, including but not limited to: heat shock transformation, electroporation, DEAE-dextran transfection, microinjection, lipid Body-mediated transfection, calcium precipitation, protoplast fusion, particle bombardment, viral transformation and similar techniques.
  • the polypeptide of interest is protected by the formation of insoluble aggregates, and the problem that the long polypeptide in the recombinant expression is easily degraded in the cell is solved; the ffi is enhanced by the solubilized peptide tag.
  • the solubility of the polypeptide of interest improves the expression of the polypeptide of interest; the self-aggregation and self-cleavage of the fusion protein enables simple separation and purification operations, avoiding expensive column separation.
  • the method of the present invention is a low cost, simple, and efficient method for the production and purification of polypeptides suitable for industrial applications.
  • Example 1 Construction of ELK16 fusion expression vector
  • the ⁇ ⁇ - lutein-ELK16 vector was first constructed, and the vector structure is shown in Fig. 1C, wherein the "Target peptide" sequence is the sequence of Bacillus subtilis lipase A (LipA).
  • the pET ⁇ OaC ⁇ vector of the commercial plasmid Novogen was selected, and the LipA polynucleotide from Bacillus subtilis lipase A was analyzed by overlapping PCR using the online tool DNAworks PT-type linker peptide and the nucleotide sequence of ELK16.
  • the ELK16 polynucleotide with PT-type spacer was synthesized in the N-terminal sequence of the LipA gene, and this polynucleotide was inserted between the Ndel and Xhol sites of the pET ⁇ 30a(+) plasmid to form pET ⁇ 30a (b). - LipA- ELK16.
  • the pET ⁇ 30a H LipA-ELKl 6 plasmid and the pTWIN1 plasmid of New England Biolab (NEB) were extracted using Tiangen's high-purity plasmid mini-kit, and the following two sets of forward and reverse primers were used, respectively, according to the conventional method.
  • PCR amplification was performed to obtain a LipA polynucleotide fragment and a Mxe GyrA intron polynucleic acid fragment:
  • Second set of primers upstream primer
  • PCR reaction using Tiangen pfo polymerase PCR reaction conditions: first 94 ° C 2min; then 94 ° C Imin, 57 ° C lmin, 72 ° C 40sec, a total of 30 cycles; the last 72 °C lOmiiio reaction after the end , 1% Joan for PCR amplification products!
  • the result of sugar gel electrophoresis showed that the correct size of the band was amplified by PCR, which was consistent with the expected results.
  • the two fragments were then separated by gel separation, and the two fragments were used as templates to carry out overlapping PCR reactions: first at 94 °C for 2 min without primers; then 94 °C Imin, 70 °C Imiii, 72 ° C 80sec, a total of 10 cycles; last 72 °C 10min.
  • the pET rx-EK-Intein-ELK16 vector was constructed, and the structure of the vector is shown in Figure ID.
  • the primers Trx-For and ⁇ - Rev in Table 1 were used to amplify the gene trxA encoding the Trx protein using the genomic UNA of the large intestine F strain BL21 (DE3), using Nde I and Spe I restriction enzymes. After digestion, the same treated pET ipA-Intein-ELK16 vector was inserted, the ligation product was transformed into E. coli BL21 (DE3) competent cells, and the transformed cells were plated with 50 g/mL kanamycin added. Positive clones were screened on LB plates, plasmids were extracted and sequenced, and the sequencing results showed that the correct pE Trx-Intein-ELK16 was obtained.
  • Trx-EK-For and Trx-EK-Rev in Table 1 and pE LipA-Intein-ELK16 as a template to amplify the intein gene with EK site and restriction endonuclease with Bgl II and Hind III
  • the enzyme was digested and inserted into the same treated pET-Trx-intein-ELK16 plasmid vector for ligation, and the ligation product was transformed into E. coli BL2i (DE3) competent cells, and the transformed cells were coated with 50 ⁇ 8 / Positive clones were screened on LB plates of ⁇ £ kanamycin, and plasmids were extracted and sequenced. The sequencing results showed that the cloned pET-Trx-EK-intein-ELK16 sequence was correct. Table 1
  • Glucagon-like peptide GLP-1 B-type natriuretic peptide BNP, prostaglandin secreted jft Ex-4, chemokine CCL5, stromal cell-derived factor SDF-ia, growth-promoting factor IGF-1 ⁇
  • the fertile hormone Lep is used as a target for recombinant production and purification by the method of the present invention. Place The polypeptide information is shown in Table 2 below: Table 2
  • GLP-1 31 aa is soluble as a fusion protein, otherwise it will degrade (Wu et al.
  • IGF- l a 70 aa inclusion body (Zhang et al., 2010) 29 30
  • Lep 346 aa inclusion body (Gertler et al., 1998) 31 32
  • nucleotide sequence encoding the above polypeptide of interest is codon-optimized (see Table 2) so that it can be better expressed in E. coli, and the corresponding coding sequence is obtained by chemical synthesis.
  • the primer sequences used for amplification of GLP 1 are shown in Table 3 below.
  • the coding polynucleotide encoding GLP-1 was amplified by PCR using the primers GLP1 F and GLP1 R in Table 3, and inserted into the expression vector shown in Figure 1 C after double digestion with Nde I and Spe I.
  • pET- LipA Intdn-ELK16 is also between the Nde I and the intein coding sequence in the intein coding sequence near the 5' end of the Spe I site (17 bases from the 5' end)
  • the sequence at the 5' end of the peptide-containing (Intein) coding sequence was unchanged; on the other hand, multiple primers were used for amplification of the primers Trx-GLP1-F and GLP1-R in Table 3, and inserted into the pET-Trx after double digestion.
  • EK-Intein-ELK16 Bgl II and Spe I sites are introduced, and the enterokinase (EK) cleavage site DDDDK (Asp-Asp-Asp-Asp- Lys) is introduced between the Bgl II site and the polypeptide of interest.
  • EK enterokinase
  • the fusion expression construct obtained in Example 2 was transformed into E. coli BL21 (DE3) competent cells by calcium chloride normalization, and positive clones were identified by colony PCR and plasmid sequencing.
  • the positive clones were ligated into LB medium, and the cells were harvested by induction of expression of 6 ho at 23 ° C, 30 ° C, and 37 ° C with 0.2 mM IPTG, and the bacterial concentration OD 6 was measured. () (The following will be 1 mL of OD 6 «. The amount of cells for 1 is called 1 OD).
  • the cells were resuspended in lysis buffer (2.4 g Tris, 29.22 g NaCl, 0,37 g Na 2 EDTA-2H 2 0 in 800 mL water, adjusted to pH 8.2, and made up to 1 L with water) To 20 OD/mL, sonication. The supernatant and the fraction were separately collected by centrifugation i0 min at 4 ° C, 10000 rpm. The precipitate was washed once with a lysis buffer supplemented with 0.5% of surfactant Triton X-100 (removal of impurities such as cell membrane fragments) and washed twice with lysis buffer to substantially remove Triton X-100.
  • lysis buffer 2.4 g Tris, 29.22 g NaCl, 0,37 g Na 2 EDTA-2H 2 0 in 800 mL water, adjusted to pH 8.2, and made up to 1 L with water
  • the washed precipitate fraction was fully resuspended with an internal guanidine cleavage buffer containing 40 mM DTT (0.62 g of dithiothreitol dissolved in 100 mL of lysis buffer and placed at - 20 ° C for use).
  • the cleavage-precipitated part of the OD cells was resuspended in a 15 mL intein-cleaving buffer (20 OD/mL) and placed under 4 ⁇ for 24 h, allowing the sputum to fully self-cleavage.
  • FIG. 2A shows the expression and purification results of the polypeptide of interest without the T « tag
  • FIG. 2B shows the expression and purification results of the polypeptidyl group with the T c tag.
  • Lane a the part of the pellet after disruption; lane b: the fusion protein is self-cleaved by indole-containing cleavage and pelleted after centrifugation; Lane c: the supernatant of the fusion protein after intein-mediated self-cleavage Part; Lane p: Sample purified by HPLC purification; Lane 1-3: Protein quantification standard (Std), where the larger band is bovine serum albumin BSA (67 kD), and the loading amount is 3 ⁇ 8 , 1.5 ⁇ , 0,75 t ag; the smaller of the bands is bacteriocin Aprotinin (6,5 kD), the loading amount is ⁇ ⁇ , 5 g, 0.75 ⁇ 8 , 0, 3 ⁇ 8 ; 4- 7 : Protein quantification standards containing only bovine serum albumin BSA, the loadings were 6 ⁇ 8 , 3 ⁇ 8 , 1.5 , and 0.75 g , respectively.
  • Std Protein quantification standard
  • the positions of the fusion protein, the cut Intein-ELK16, and the target 3 ⁇ 4 are indicated by the legend.
  • the molecular weights of the molecular weight standards Ml (14 ⁇ 94 kD) and M2 (3.3 ⁇ 20.1 kD) correspond to the molecular weights listed in the protein.
  • the glue picture is on the left and right side.
  • Bio-Rad's Quantity ONE gel quantitative analysis software is used to perform optical density analysis on the strip, which can be used to calculate the aggregate yield of the fusion protein, after intein-mediated self-cleavage.
  • the yield of the polypeptide of interest released into the supernatant, and the purity of the polysaccharide in the supernatant, are shown in Table 4.
  • polypeptides of interest in the seven types of medium-length polypeptides namely GLP1, CCL5, SDF-1 ⁇ , IGF-1 and Lep, which can form a large number of aggregates after fusion with Intein ELK16, and the expression amount is 28.4-44.6 g. /mg cell wet weight.
  • the target polypeptides BNP and Ex 4 were not given in Fig. 2 because no foreign protein expression was observed in the supernatant and the precipitate after disruption of the cells.
  • the target polypeptide GLP-1 can be directly released into the supernatant after intein-mediated cleavage, and the recovery rate is about 46.8 %, and the yield is 1. 8 g/mg cell wet weight. Therefore, the target of the supernatant, GLP-1, was directly purified by reverse phase HPLC. As shown in Fig. 2, the purity of the final product was over 95%, and the final yield was about 0.8 g/mg of cell wet weight.
  • IGFl o SDF-1 alpha, CCL-5 and Leptin and lutein-ELK16 fusion expression are mostly distributed in aggregates, they are insoluble precipitates after self-cleavage by indole-containing cleavage.
  • the target polypeptides GLP-1, BNP, Ex-4, CCL5, SDF-1a and IGF-1 ⁇ obtained by fusion with Trx obtained in Example 3 were further isolated and purified.
  • enterulin kinase Enterokinase, New England BioLabs, Cat. No. P8070S
  • enterulin kinase was used to cleave the fusion protein Trx- ⁇ released into the supernatant after intein-mediated self-cleavage. The specific steps are:
  • Trx tag and the polypeptide of interest generated after enterokinase cleavage can be further purified by reverse phase HPLC.
  • the detailed procedure is as follows:
  • the black arrow indicates the ⁇ label produced by enterokinase cleavage
  • the blue arrow indicates the polypeptide of interest produced by enterokinase cleavage.
  • the molecular weights of the molecular weight standards Ml (14 ⁇ 66 kD) and M2 (3,3 ⁇ 20.1 kD) are respectively listed on the left and right sides of the protein gel image.
  • the target band is subjected to optical density analysis using Bio Rad Quantity ONE, and the yield and purity of the target polypeptide in the sample are further calculated.
  • Trx-tagged GLP1, BNP, Ex4 and SDF-lot can be efficiently recovered and purified by reverse phase HPLC, and the final product purity is over 95%, and the final yield is about 0.3-L8 g/mg cell wet weight. See Table 5 below for the specific results:
  • solubilizing the peptide tag may be a combination of self-assembling peptide
  • the self-aggregating peptide can mediate the formation of insoluble aggregates of the fusion protein, and is suitable for rapid separation and purification by methods such as centrifugation or filtration.
  • a lytic peptide tag is added to improve expression of the polypeptide of interest and to increase the solubility of the polypeptide of interest.
  • Trx In addition to Trx, a selection of the other five commonly used solubilizing labels in the table below was used for peptide production and purification in combination with self-aggregation.
  • the fusion proteins SUMO-thymosin (Thymosm)/BNP-intein-ELKl 6 , GST-GLP 1 -intein-ELKl 6 , DsbC- GLPi- intein- ELKi6, GBi- GLPi were constructed respectively.
  • Trx-GLP1-intein-18A, Trx-GLP1-intein-L6KD Two other self-aggregating peptides, 8A and L6KD, which have similar functions to ELK16, were also tested.
  • the expression vectors of the fusion proteins Trx-GLP1-intein-18A, Trx-GLP1-intein-L6KD were constructed according to the methods described in the above examples, and recombinant expression and separation based on self-aggregating peptides were performed according to the methods described above. purification.
  • Gertler, et al. Large-scale preparation of biologically active recombinant ovine obese protein (leptin). FEBS Letters 422 (1998) 137-140.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Endocrinology (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

公开了多肽的产生和纯化方法,具体公开了包含促溶肽标签、自聚集肽和目的多肽的融合蛋白,以及通过表达所述融合蛋白来产生和纯化目的多肽的方法。

Description

多) ¾的产生和纯化方法 技术领域
本发明涉及基因工程领域。 更具体地, 本发明涉及包含促溶肽标签部 分、 自聚集肽部分和目的多肽部分的融合蛋白和通过表达所述融合蛋白来 产生和纯化目的多肽的方法。 说 背景技术 月 目前, 关于多牍在医药中应用的研发已广泛涉及抗肿瘤药物、 心脑血 管药物、疫苗和抗病毒药物, 以及诊断试剂盒等多个方面 (Leader等, 2008)。 同迅速增长的市场需求相比, 多肽的生产方法在一定程度上限制了其发展。 常规的化学固相合成法在生产超过 30个氨基酸的中长多肽时, 随着肽段长 度增大, 合成的成本和难度将大幅增加 (Bray等, 2003)。
另一种有效的手段是采用重组方法在宿主细胞内产生多駄。 利用大肠 杆菌生长速率快、 表达量高、 生产成本低等特点, 通过引入外源基因, 或 者操作基因改变氨基酸序列, 能够很容易生产得到所需的多駄产品, 而且 该方法操作步骤简便、 易于放大生产。 目前市场上超过 30%的重组多肽类 药物是使用大肠杆菌细胞生产的 (Kamionka等, 2011 ; Demain等, 2009; Walsh, 2003和 2006)。 然而, 长度小于 100个氨基酸的短肽很容易被蛋白 醸降解, 大大影响产率 (Murby等, 1996; Kuliopulos等, 1994; Hamiig等, 1998)。
在重组产生多牍的方法中, 纯化步骤非常关键。 据报道, 重组多) ¾的 分离与纯化成本约占其全部生产成本的 60%- 80% (陈浩等人, 2002)。 重组 多肽的纯化方法包括传统的离子交换层析、 疏水性相互作用层析、 亲和层 析等。 离子交换层析和疏水性相互作用层析由于对样品起始条件育一定的 要求, 通用性和效率不及亲和层析。 亲和纯化通常可以获得超过 90%的高 收率, 使其成为目前最常用的重组蛋白纯化方法。 常用的亲和纯化技术包 括组氨酸标签 (his- tag)或谷胱甘肽转移酶标签 (GSTTtag)与目的多肽的融合 表达, 为不同目的多駄的生产提供了通用的纯化手段。 然而昂贵纯化柱使 亲和纯化成本较高, 不利于工业领域的应用。
本领域仍需要低成本、 简便、 高效的多牍产生和纯化方法。 发明概述
本发明提供了基于自聚集肽和促溶牍标签的低成本、 简便、 高效的多
)¾产生和纯化方法。
在第一方面, 本发明提供了一种融合蛋白, 其包含促溶) ¾标签部分、 自聚集肽部分以及目的多肽部分, 其中所述目的多肽部分位于所述促溶肽 标签部分和所述自聚集肽部分之间, 并且所述目的多肽部分通过第一接头 连接于所述自聚集肽部分, 其中所述第一接头包含第一切割位点, 所述融 合蛋白在宿主细胞内表达后 通过所述自聚集肽部分形成活性聚集体。 在 一些实施方案中, 所述自聚集肽部分位于所述融合蛋白的 C端。
在一些实施方案中, 所述自聚集肽部分包含两亲性自组装短肽。 在一 些实施方案中, 所述两亲性自组装短肽选自两亲性 β折叠短肽、 两亲性 α 螺旋短肽、 类表面活性剂短肽。
在一些实施方案中, 所述两亲性 β折叠短肽的长度为 4-30个氨基酸残 基。 在一些实施方案中, 所述两亲性 β折叠短肽的疏水性氨基酸残基含量 为 40%-80%。 在一具体实施方案中, 所述两亲性 β折叠短肽的氨基酸序列 示于 SEQ ID NO 在一些实施方案中, 本发明的融合蛋白中的自聚集肽 部分包括 1个所述两亲性 β折叠短肽。 在另一些实施方案中, 本发明的融 合蛋白中的自聚集肽部分包括串联重复的两个或更多个所述两亲性 β折叠 短肽。
在一些实施方案中, 所述两亲性 α螺旋短肽的长度为 4-30个氨基酸残 基。 在一些实施方案中, 所述两亲性 α螺旋短肽的疏水性氨基酸残基含量 为 40%-80%。 在一具体实施方案中, 所述两亲性 α螺旋短肽的氨基酸序列 示于 SEQ ID NO 在一些实施方案中, 本发明的融合蛋白中的自聚集肽 部分包括 1个所述两亲性 α螺旋短肽。 在另一些实施方案中, 本发明的融 合蛋白中的自聚集肽部分包括串联重复的两个或更多个所述两亲性 α螺旋 短肽。
在一些实施方案中, 所述类表面活性剂短肽具有 7-30个氨基酸残基, 其从 Ν端到 C端具有以下通式表示的氨基酸序列: A-B或 B-A
其其中中 AA是是亲亲水水性性氨氨基基酸酸残残基基组组成成的的肽肽,, 所所述述亲亲水水性性氨氨基基酸酸残残基基可可以以是是 相相同同的的或或不不同同的的,, 且且选选自自 LLyyss、、 AAsspp.. AArrgg、、 GGlluu、、 HHiiss.. SSeerr、、 TThhrr、、 AAssnn和和 GGiinn;;
BB 是是由由疏疏水水性性氨氨基基酸酸残残基基组组成成的的肽肽,, 所所述述疏疏水水性性氨氨基基酸酸残残基基可可以以是是相相同同的的 55 或或不不同同的的,, 且且选选自自 LLeeuu、、 GGllyy、、 AAllaa.. VVaall、、 IIllee、、 PPhhee和和 TTiipp;; 且且 AA与与 BB通通过过 肽肽键键连连接接;; 并并且且其其中中在在所所述述类类表表面面活活性性剂剂短短肽肽中中疏疏水水性性氨氨基基酸酸残残基基的的比比例例 是是 5555%%--9955%%。。 在在一一些些实实施施方方案案中中,, 所所述述类类表表面面活活性性剂剂短短肽肽具具有有 88个个氨氨基基酸酸 残残基基,, 其其中中疏疏水水性性氨氨基基酸酸残残基基的的比比例例是是 7755%%。。 在在一一些些具具体体的的实实施施方方案案中中,, 所所述述类类表表面面活活性性剂剂短短肽肽的的氨氨基基酸酸序序列列示示于于 SSEEQQ IIDD NNOO::33、、 SSEEQQ IIDD NNOO::44或或 1100 SSEEQQ IIDD NNOO::55。。
在在一一些些实实施施方方案案中中,, 本本发发 的的融融合合蛋蛋白白中中的的促促溶溶肽肽标标签签选选自自 NNuussAA、、 GGSSTT、、 TTrrxx、、 SSUUMMOO,, DDssbbCC、、 ZZ、、 GGBB11、、 MMBBPP禾禾 ΠΠ TT77PPKK。。
在在一一些些实实施施方方案案中中,, 本本发发明明的的融融合合蛋蛋白白中中的的所所述述第第一一切切割割位位点点选选自自化化 学学切切割割位位点点、、 酶酶法法切切割割位位点点和和自自切切割割位位点点。。 在在一一些些实实施施方方案案中中,, 所所述述自自切切 1155 割割位位点点为为内内含含駄駄iinnteteiinn))。。 在在一一些些具具体体的的实实施施方方案案中中,, 所所述述内内含含肽肽为为序序列列示示 于于 SSEEQQ IIDD NNOO::66的的 MMxxee GGyyrrAA。。 在在一一些些具具钵钵的的实实施施方方案案中中,, MMxxee GGyyrrAA直直接接 连连接接于于所所述述目目的的多多肽肽的的 CC端端。。
在在一一些些可可选选的的实实施施方方案案中中,, 本本发发明明的的融融合合蛋蛋白白中中的的所所述述第第一一接接头头还还包包 含含间间隔隔物物。。
2200 在在一一些些实实施施方方案案中中,, 本本发发明明的的融融合合蛋蛋白白中中的的所所述述目目的的多多駄駄通通过过第第二二接接 头头连连接接于于所所述述促促溶溶肽肽标标签签。。 在在一一些些可可选选的的实实施施方方案案中中,, 所所述述第第二二接接头头包包含含 间间隔隔物物。。 在在另另一一些些可可选选的的实实施施方方案案中中,, 所所述述第第二二接接头头包包含含第第二二切切割割位位点点,, 其其中中第第二二切切割割位位点点的的切切割割条条件件不不^^于于所所述述第第一一切切割割位位点点的的切切割割条条件件。。 在在一一 些些实实施施方方案案中中,, 所所述述第第二二切切割割位位点点选选自自第第二二化化学学切切割割位位点点、、 第第二二酶酶法法切切割割
2255 位位点点和和第第二二自自切切割割位位点点。。 在在一一些些具具体体的的实实施施方方案案中中,, 所所述述第第二二酶酶法法切切割割位位 点点包包含含 SSEEQQ IIDD NNOO::77所所示示的的肠肠激激酶酶识识别别序序列列。。在在另另一一些些具具体体的的实实施施方方案案中中,, 所所述述肠肠激激酶酶识识别别序序列列直直接接连连接接于于所所述述目目的的多多肽肽的的 NN端端。。 在在一一些些可可选选的的实实施施 方方案案中中,, 所所述述第第二二接接头头除除了了第第二二切切割割位位点点外外还还包包括括间间隔隔物物。。
在在一一些些实实施施方方案案中中,, 本本发发明明的的融融合合蛋蛋白白中中的的所所述述目目的的多多肽肽为为长长度度为为
Figure imgf000004_0001
在在另另一一方方面面,, 本本发发明明提提供供了了一一种种多多核核苷苷酸酸,, 其其包包含含编编码码本本发发明明上上述述的的 融合蛋白的核苷酸序列或其互补序列。
在另一方面, 本发明提供了一种表达构建体, 其包含本发明上述的多 核苷酸。
在另一方面, 本发明提供了一种宿主细胞, 其包含本发明上述的多核 苷酸或以本发明的表达构建体转化, 其中所述宿主细胞能够表达本发明上 述的融合蛋白。
在另一方面, 本发明提供了一种产生和纯化目的多肽的方法, 所述方 法包括以下步骤:
(a) 培养本发 上述的宿主细胞, 从而表达本发明的融合蛋白;
(b)裂解所述宿主细胞, 然后去除细胞裂解物的可溶部分, 回收不溶部 分;
(c) 通过切割第一切割位点从所述不溶部分释放可溶的带有促溶駄标 签的目的多駄; 和
(d) 去除步骤 W中的不溶部分, 回收含有所述目的多肽的可溶部分。 在一些实施方案中, 本发明的产生和纯化目的多肽的方法还包括:
(e) 如果所表达的融合蛋白存在第二切割位点, 则通过切割第二切割位 点使所述目的多肽和所述促溶肽标签分离;
(f) 去除所述促溶肽标签, 获得纯化的目的多肽。 附图说明
图 1. 基于自聚集肽 (ELK16)和促溶肽标签 (Τπ 的多駄产生和纯化策略 以及表达载体图谱。 Α : 带促溶肽标签; Β : 不带促溶肽标签; pET-Lip A-Intein-ELK 16载体; D: pET Trx-EK- lutein- ELK16载体。
图 2. 目的多肽的表达与纯化结果。 A: 不带 Trx标签; 带 Trx标签。 图 3. 目标蛋白 GLP- 1与 Trx分离的 HPLC吸收峰图谱。
图 4. 融合蛋白 Trx-目的多肽- lutein- ELK16 经肠激酶切割的结果的 SDS- PAGE分析。
图 5. 促溶) ¾标签 SUMO与自聚集) ¾ ELK 16组合用于产生和纯化目的 多肽的结果。
图 6. 促溶肽标签 GST与自聚集肽 ELK16组合用于产生和纯化目的多 牍的结果。 图 7. 促溶 J¾标签 DsbC与自聚集肽 ELK16组合用于产生和纯化目的多 馱的结果。
图 8. 促溶牍标签 GB1与自聚集肽 ELK16组合用于产生和纯化目的多 肽的结果。
图 9. 促溶) ¾标签 Z与自聚集肽 ELK16组合用于产生和纯化目的多) ¾ 的结果。
图 10. 自聚集肽 18A和 L6KD与促溶牍标签 Tnc组合用于产生和纯化 GLP1多肽的结果。 发明详述
在第一方面, 本发明提供了一种融合蛋白, 其包含促溶駄标签部分、 自聚集肽部分以及目的多肽部分, 其中所述目的多肽部分位于所述促溶肽 标签部分和所述自聚集肽部分之间, 并且所述目的多肽部分通过第一接头 连接于所述自聚集肽部分, 其中所述第一接头包含第一切割位点, 所述融 合蛋白在宿主细胞内表达后可通过所述自聚集肽部分形成活性聚集体。
在第二方面, 本发明提供一种多核苷酸, 其包含编码本发明的融合蛋 白的核苷酸序列或其互补序列。
在第三方面, 本发明提供一种表达构建体, 其包含本发明的多核苷酸。 在第四方面, 本发明提供一种宿主细胞, 其含有本发明的多核苷酸或 以本发明的表达构建体转化, 其中所述宿主细胞能够表达本发明的融合蛋 白。
在第五方面, 本发明提供一种产生和纯化目的多肽的方法, 所述方法 包括以下步骤: (a) 培养本发明的宿主细胞,从而表达所述融合蛋白; (b)裂 解所述宿主细胞, 然后去除细胞裂解物的可溶部分, 回收不溶部分; (c)通 过切割第一切割位点从所述不溶部分释放可溶的带有促溶肽标签的目的多 肽; 和 (d) 去除步骤 (C)中的不溶部分, 回收含有所述目的多肽的可溶部分。
如本文所用, 术语 "多) ¾ "和 "蛋白"可互换使用, 并且定义为由通 过肽键连接的氨基酸残基组成的生物分子。如本文所用, " 的多駄 "或"目 的蛋白" 是指可通过本发明的方法产生并纯化的任何多肽或蛋白质, 其非 限制性例子包括酶、 激素、 免疫球蛋白链、 诸如抗癌多肽的治疗性多) ¾、 诊断性多) ¾或者可以用于免疫 §的的多) ¾或其生物学活性片段等等。 目的 多肽可以来自任何来源, 包括微生物来源多肽、 喃乳动物来源多肽和人工 蛋白质 (例如融合蛋白或突变的蛋白质)等等。
目的多肽可以是任何长度的多肽和蛋白。 可通过本发明的方法产生并 纯化的目的多肽的长度可以是 20-200, 25-150, 30-120, 30- 100个氨基酸残 基, 例如, 大约 30, 大约 40, 大约 50, 大约 60, 大约 70, 大约 80, 大约 90个氨基酸残基。
可通过本发明的方法来产生并纯化的 "目的多駄" 的实例包括但不限 于胰高血糖素样肽 GLP- i、 B型脑利钠肽 BNP、 促姨岛素分泌肽 Ex- 4、 趋 化因子 CCL5、 基质细胞衍生因子 SDF- 1α、 促生长因子 IGF- ia、 肥胖荷尔 蒙 Lep、 P条丐素 (Calcitonin)、 ' 莫 -瑞'林 (Sermordin)、 '胸,腺 H太 (Thymosin)、 ;X 蛭素 (Lepimdin)、天査素 (Cecropin)、人富组蛋白 (Histatin)、防御素 (defensin)、 和人纤溶酶原 (Kringle i- 5)或它们的生物学活性片段等。
如本文所用, "促溶肽标签"是指与目的多肽融合后能够帮助目的多駄 正确折叠并提高融合蛋白可溶性的融合标签。 本领域技术人员已知 多这 样的 "促溶肽标签"。 可用于本发明的合适的促溶肽标签包括但不限于
NusA、 GST、 Trx、 SUMO , DsbC、 Z、 GB1 , MBP禾口 T7PK(Leder et al, 2007; Esposito和 Chatteijee, 2006; Waugh, 2005)。在一个优选实施方案中, 所述促 溶肽标签为 Trx。
如本文所用, "自聚集肽"是指与目标多肽部分融合并在宿主细胞表达 后能够介导融合蛋白在胞内形成不可溶的活性聚集体的多肽。 如本文所用,
"活性聚集体"指的是目标多肽部分仍然能够正确折叠并保持活性或者是 指聚集体中的目的多肽部分在与自聚集駄分离后能够处于可溶状态。
无意于受到任何理论的限制, 本领域中已知一些两亲性 (amphipathic) 多肽由于具有彼此分隔的亲水性区域和疏水性区域, 在疏水相互作用以及 其它推动力作用下能自发地形成特定的自组装结构 (Zhao 等, 2008)。 本发 明人令人惊奇地发现一些具有自组装能力的两亲性短肽能够诱导细胞内活 性聚集体的形成。 用作本发明的自聚集肽的两亲性自组装短肽可以选自两 亲性 β折叠短肽、 两亲性 α螺旋短肽和类表面活性剂短肽。
如本文所用, "两亲性 β折叠短肽"是指具有 4-30个氨基酸残基, 由疏 水性氨基酸、 带电荷的亲水性氨基酸交替排列构成的短肽, 其形成 β折叠 时, 一侧为疏水氨基酸残基, 另一侧是交替排列带正电荷和带负电荷的亲 水性氨基酸残基。 这些短肽可以在疏水相互作用、 静电相互作用以及氢键 作用下形成自组装结构。 一般而言, 两亲性 β折叠结构的长度越长或疏水 性越强, 自组装越容易发生, 形成的自聚集体的机械强度越强。 为了保证 足够的自组装能力, 本发明的两亲性 β折叠短肽应当包含一定量的疏水性 氨基酸。 本发明的两亲性 β折叠短肽包括 40-80%、 45-70%、 50-60%, 例如 大约 50%的疏水性氨基酸残基。 可用作本发明的自聚集肽的两亲性 β折叠 短肽的具体实例是氨基酸序列示于 SEQ ID ΝΟ: 1的 ELK16 (氨基酸序列: LELELKLKLELELKLK)。
本领域已有通过多个重复单元串联重复形成具有自聚集特性的多肽的 报道, 如类弹性蛋白 ELP, 其由 110个 VPGXG重复单元组成, 其聚集特 性与重复单元数目相关 (Banki, et al., 2005; MacEwan和 Chilkoti, 2010)。也有 报道显示由多个重复单元组成的两亲性 β折叠的自聚集倾向随着重复单元 数目增加而增强 (Zhang et al, 1992)。 可以预期, 由多个上述 "两亲性 β折 叠短肽" 串联组成的多肽能够保留甚至获得增强的自组装能力。
因此, 本发明的自聚集肽部分可以包括一或多个串联连接的所述两亲 性 β折叠短肽。本发明的自聚集肽部分可以包含 1-150、 1-130、 1-110、 1-90、 1-70、 1-50、 1-30、 1-10个、 1-5个, 例如 1、 2、 3、 4、 5个所述两亲性 β 折叠短肽。 所述自聚集肽部分中的两或多个两亲性 β折叠短肽可以形成串 联重复。 为了便于重组操作以及考虑到生产成本问题, 期望使用较少的重 复。 因此, 在一些实施方案中, 所述 "自聚集肽部分"仅包含一个所述两 亲性 β折叠短肽。
α螺旋是肽链骨架围绕一个轴以螺旋的方式伸展的一种蛋白二级结构。 如本文所用, "两亲性 α螺旋短肽"是指具有 4-30个氨基酸残基, 与普通 α 螺旋相比具有独特的亲水、 疏水氨基酸排列, 使得在形成的 α螺旋结构的 一侧主要为亲水性氨基酸, 而在另一侧主要为疏水性氨基酸的短肽。 据推 测两亲性 α螺旋在水溶液中通过形成卷曲螺旋 (coiled-coil)而实现自组装, 其中两个 α螺旋通过疏水相互作用结合, 并进一步通过带电氨基酸的静电 相互作用力稳定这种结合。 本发明的两亲性 α 螺旋短肽包括 40-80%、 45-70%、 50-60%, 例如大约 50%的疏水性氨基酸残基。 可用作本发明的自 聚集肽的两亲性 α螺旋短肽的具体实例为氨基酸序列示于 SEQ ID NO:2的 18 A (EWLKAFYEKVLEKLKELF)。 与两亲性 β折叠类似, 由两亲性 α螺旋短肽串联组成的多肽保持甚至 增强自组装能力。 本发明的 "自聚集肽部分"可以包括一或多个串联连接 的所述两亲性 α螺旋短肽。例如,本发明的"自聚集肽部分 "可以包含 1-150、 1-130、 1-110、 1-90、 1-70、 1-50、 1-30、 1-10个、 1-5个, 例如 1、 2、 3、 4、 5个所述两亲性 α螺旋短肽。所述自聚集肽部分中的两或多个两亲性 α螺旋 短肽可以形成串联重复。 不过, 为了便于重组操作以及考虑到生产成本问 题, 期望使用较少的重复。 因此, 在一些实施方案中, 本发明的 "自聚集 肽部分"仅包含一个所述两亲性 α螺旋短肽。
"类表面活性剂肽"是可用作本发明的自聚集肽的另一类两亲性多肽, 其通常由 7-30个氨基酸残基组成, 延伸长度约 2-5nm, 结构类似于脂质, 由一段疏水性氨基酸尾部和亲水性氨基酸头部构成。 类表面活性剂结构的 性质类似于表面活性剂, 在水溶液中可以形成胶束、 纳米管等组装结构。 适于用作本发明的自聚集肽的类表面活性剂短肽的长度可以是 7-30个氨基 酸残基, 其包括从 N端到 C端的以下通式表示的氨基酸序列:
A-B或 B-A,
其中 A与 B之间通过肽键连接。 A是由亲水性氨基酸组成的亲水性头部, 所述亲水性氨基酸残基可以是相同的或不同的, 且选自 Lys、 Asp, Arg、 Glu、 His. Ser、 Thr、 Asn和 Gln。 A的实例包括 KD、 KK等。 B是由疏水 性氨基酸残基组成的疏水性尾部, 所述疏水性氨基酸残基可以是相同的或 不同的,且选自 Leu、Gly、Ala、 Val、Ile、Phe和 Trp。B的实例包括 LLLLLL(L6) 或 GAVIL等。 本发明的类表面活性剂短肽中疏水性氨基酸比例高于亲水性 氨基酸的比例, 在所述类表面活性剂短肽中的疏水性氨基酸比例可以是 55-95%, 60-95%, 65-95%, 70-95%, 75-95%, 80-95%, 85-95%, 90-95%。 在一些实施方案中, 所述类表面活性剂短肽具有 8个氨基酸残基, 其中疏 水性氨基酸的比例是 75%。 适用于本发明的自聚集肽的类表面活性剂短肽 的具体实例包括氨基酸序列分别示于 SEQ ID NO:3、 SEQ ID NO:4或 SEQ ID NO: 5的 L6KD、 L6K2或 DKL6。
此外, 巳有报道, 一些蛋白结构域, 例如 β淀粉样肽、 VP1、 MalE31、 CBDds等, 也能够诱导融合蛋白形成聚集体, 本发明预期这样的结构域也 可用作本发明 "自聚集肽"。 然而这些结构域的结构相对复杂并且其诱导聚 集的机理伋不清楚 (Mitraki, 2010)。 在本发明中优选使用结构相对简单以及 长度较短的两亲性自组装短肽。
本发明人之前的研究已经发现, 具有诱导活性聚集体形成的能力的自 聚集肽 (如两亲性自组装肽 与目的多肽作为融合蛋白在宿主细胞内表达后, 表达的融合蛋白可形成不可溶的聚集体。 聚集体的形成可以避免胞内蛋白 酶对融合蛋白的降解, 因此提高目的多牍的产量。 细胞裂解后, 可以简单 地通过离心沉淀或过滤等方法从细胞裂解物中收集不溶的聚集体, 除去可 溶的杂质, 实现对融合蛋白的初步纯化。 之后, 通过切割位于自聚集牍部 分以及目的多肽之间的接头中的切割位点, 使得可溶的包含目的多肽的部 分从不可溶部分 (沉淀)释放出来, 分布于上清液中, 再次简单地通过离心沉 淀或过滤等方法即可去除不溶的杂质, 收获可溶的目的多肽。 通过这样的 基于自聚集駄的方法生产多肽 (示意图见于图 1A)可以筒化分离纯化步骤, 避免使用昂贵的纯化柱, 显著地降低生产成本。
本领域中已知促溶肽标签可以提高融合蛋白的可溶性, 这与自聚集駄 倾向于使融合蛋白不可溶的作用相反。 然而, 本发明人令人惊奇地发现, 当包含促溶肽标签部分、 目的多肽部分和自聚集牍部分的融合蛋白在宿主 细胞内表达后仍能形成不溶聚集体, 仍然可以通过上述步骤容易地进行分 离纯化 (示意图见于图 1B), 而促溶肽标签可以进一步促进目的多牍的正确 折叠, 还可以在去除自聚集肽部分后保持或提高目的多肽的可溶性。 无意 于受到任何理论的限制, 这种结果据推测是由于促溶肽标签和自聚集駄能 够形成某种功能上的平衡。 此外, 本发明人还发现促溶肽标签的使用还可 以改善融合蛋白的表达, 可用于生产某些原来难以重组生产的多肽。
根据本发明, 目的多肽通过第一接头连接于所述自聚集駄部分, 其中 所述第一接头包含第一切割位点。 如本文所用, "切割位点"包括实现切割 所需的序列, 如用于酶法切割的蛋白醇识别序列、 用于自切割的内含馱序 列等。
本发明的用于将可溶的包含目的多肽的部分从不可溶部分 (沉淀)释放 出来的第一切割位点包括可以化学切割、 酶法切割或自切割的切割位点, 或本领域技术人员已知的其它任何切割位点。 本发明中优选的第一切割位 点可以进行自切割, 例如, 其包含可自切割的内含肽的氨基酸序列。 这是 因为基于内含肽的切割方法不需要外加酶或使用如化学法中所用的溴化氢 等有害物质, 而仅仅需要改变聚集体所处的缓冲环境就能筒单地诱导切割 (Wu et al., 1998; TELENTI et al, 1997)。 本领域已知多种自切割内含駄, 例 如 NEB公司的一系列具有不同自切割特性的内含肽。在一个具体的实施方 案中, 所述内含肽为序列示于 SEQ ID NO:6 的 Mxe GyrA, 通过在缓冲体 系中加入合适量的二硫苏糖醇 就可诱导该内含肽在其羧基端的自切 本发明的融合蛋白中目的蛋白可以与促溶肽标签直接连接, 或者可以 通过第二接头与促溶牍标签连接。 如果需要获得不带标签的目的多肽, 可 以在第二接头中再引入第二切割位点。 在进行第一次切割步骤使得目的多 駄与自聚集肽部分分离后, 通过切割第二切割位点, 进一步将目的多駄和 促溶肽标签分离, 再通过进一步的分离纯化 (如通过 HPLC), 获得不带标签 的目的多肽。因此,本发明的产生和纯化目的多駄的方法还可以包括: (d) 如 果存在第二切割位点, 则通过切割第二切割位点使目的多肽和促溶肽标签 分离; (e) 去除所述促溶駄标签, 获得纯化的目的多駄。
本发明的 于将目的多駄和促溶肽标签分离的第二可切割位点包括可 以化学切割、 酶法切割或自切割的切割位点, 或本领域技术人员已知的其 它任何切割位点。 在一些实施方案中, 所述第二切割位点为酶法切割位点。 在一些具体的实施方式中, 所述酶法切割位点包含肠激酶识别位点 (氨基酸 序列: DDDDK, SEQ ID NO:7) o
应当理解, 所述第二切割位点应具有与第一切割位点不同的切割条件, 使得在切割第一切割位点时第二切割位点不被切割, 目的蛋白仍保持与促 溶肽标签相连接。 这可以通过多种方式来实现, 非限制性实例包括, 例如, 第一切割位点为自切割而第二切割位点通过酶法切割, 或者第一和第二切 割位点均为酶法切割但是分别通过不同的酶来切割等等。
本领域技术人员能够理解, 为了减少本发明的融合蛋白中不同部分之 间的相互干扰,可以通过间隔物连接融合蛋白的不同部分。如本文所用, "间 隔物"是指具有一定长度的由低疏水性和低电荷效应的氨基酸组成的多肽, 其用于融合蛋白 可以使所连接的各部分充分展开, 互不干扰地充分折叠 成各自的天然构象。 因此, 所述第一接头和 /或第二接头除切割位点外还可 以额外包含间隔物。 在一些实施方案中, 如无需除去促溶肽标签, 本发明 的融合蛋白中的第二接头可以仅包含间隔物而不含切割位点。
本领域常用的此类间隔物包括例如, 富含甘氨酸 (G)和丝氨酸 (S)的柔性 的 GS型间隔物; 富含脯氨酸 (P)和苏氨酸 0 的刚性的 PT型间隔物。 由于 PT型间隔物通常相对于 GS型间隔物具有更好的蛋白醸耐受性, 因而在本 发明中优选使用 PT型间隔物。在一些具体实施方案中, 本发明所使用的间 隔物是序歹为 PTPPTTPTPPTTPTPT(SEQ ID NO:8)的 PT型间隔物。
在多駄类药物的生产中, 常常需要重组产生的多肽与目的多肽具有一 致的序列, 即两端不具有额外的氨基酸残基。 在本发明中, 这可以通过选 择合适的第一和第二切割位点及它们与 的多肽的连接方式来实现。 本领 域技术人员清楚如何根据切割位点的特性来进行这样的选择。 例如, 在一 个具体实施方案中,可以使第一切割位点的 Mxe GyrA与所述目的多駄的 C 端直接连接,使得其与所述目的多駄之间没有额外的氨基酸残基。由于 Mxe GyrA直接在其 N端进行切割, 这样经切割产生的目的多肽 C端没有多余 的氨基酸残基。 在另一个具体实施方案中, 可以使第二切割位点的肠激酶 识别位点与所述目的多駄的 N端直接连接, 使得其与所述目的多肽之间没 有额外的氨基酸残基。 由于肠激酶直接在其识别位点 C端进行切割, 这样 最后获得的目的多肽 N端没有多余的氨基酸残基。 如果要获得两端均不含 任何多余氨基酸残基的目的多牍, 则可通过例如, 使第一切割位点的 Mxe GyrA与所述目的多駄的 C端直接连接并且使第二切割位点的肠激酶识另 ί]位 点与所述目的多肽的 Ν端直接连接来实现。
如上所述, 本发明也涉及多核苷酸, 其包含编码本发明的融合蛋白的 核苷酸序列或其互补序列。 如本文所用, "多核苷酸"是指多个核苷酸通过 3'-5'-憐酸二酯键连接而成的大分子, 其中所述核苷酸包括核糖核苷酸和脱 氧核糖核苷酸。 本发明的多核苷酸的序列可以针对不同的宿主细胞 (如大肠 杆菌)进行密码子优化, 从而改善融合蛋白的表达。 进行密码子优化的方法 是本领域已知的。
如上所述, 本发明也涉及包含本发明上述的多核昔酸的表达构建俥。 在本发明的表达构建体中, 编码所述融合蛋白的多核苷酸的序列与表达控 制序列可操纵地连接以进行希望的转录及最终在宿主细胞中产生所述融合 蛋白。 合适的表达控制序列包括但不限于启动子、 增强子、 核糖体作用位 点如核糖体结合位点、 聚腺苷酸化位点、 转录剪接序列、 转录终止序列和 稳定 mRNA的序列等等。
用于本发明的表达构建体的载体包括那些在宿主细胞中自主复制的载 体, 如质粒载体; 还包括能够整合到宿主细胞 DNA中并和宿主细胞 DNA 一起复制的载体。 可商购获得许多适于本发明的载体。 在一个具体实施方 案中, 本发明的表达构建体衍生自 Novagen公司的 pET30a(+)。
本发明还涉及一种宿主细胞, 其含有本发明的多核苷酸或以本发明的 表达构建体转化, 其中所述宿主细胞能够表达本发明的融合蛋白。 用于表 达本发明融合蛋白的宿主细胞包括原核生物、 酵母和高等真核细胞。 示例 性的原核宿主包括埃希氏菌属 ( c/ien'di i 、 芽孢杆菌属 ( β«7ΖΜ 、 沙门氏 菌属 (5^wi。《e〃a)以及假单胞菌属 ( ^£?Μί/。《ί。《ίΜ)禾口链霉菌属 (Sireptomyci^)的 细菌。 在优选的实施方案中, 宿主细胞是埃希氏菌属细胞, 优选是大肠杆 菌。 在本发明的一个具体实施方案中, 所使用的宿主细胞为大肠杆菌 BL21 (DE3)菌株细胞 (Novagen)。
可以通过许多已熟知的技术之一将本发明的重组表达构建体导入宿主 细胞, 这样的技术包括但不限于: 热激转化, 电穿孔, DEAE-葡聚糖转染, 显微注射, 脂质体接介导的转染, 憐酸钙沉淀, 原生质融合, 微粒轰击, 病毒转化及类似技术。
本发明上面描述的用于生产和纯化目的多肽的方法中, 通过形成不溶 聚集钵保护目的多肽, 解决了重组表达中长多肽在胞内易被降解的问题; 通过促溶肽标签的使 ffi增强了目的多肽的可溶性, 改善了目的多肽的表达; 融合蛋白的自聚集和自切割使得可以实现简单的分离纯化操作, 避免了昂 贵的柱分离。 因此, 本发明的方法是一种低成本、 简便、 高效的适于工业 应用的多肽产生和纯化方法。 实施例
下面将通过实施例的方式进一步说明本发明, 但并不因此将本发明限 制在所描述的实施例范围中。 实施例 1: 构建 ELK16融合表达载体
本申请实施例中所使用的表达载体 pET~LipA- Inteiri- ELK16 和 pET-Trx-EK-Intein-ELKl 6的构建过程如下:
首先构建 ρΕΤ ϋρΑ- lutein- ELK16载体, 该载体结构如图 1C所示, 其 中 "Target peptide"序列为枯草芽孢杆菌脂肪酶 A (LipA)的序列。 选择商用质粒 Novogen公司的 pET^OaC^载体,用在线工具 DNAworks 设什 PT型连接肽和 ELK16的核苷酸序列, 利用重叠 PCR的方法将来自枯 草芽抱杆菌脂肪酶 A的 LipA多核苷酸和带 PT型间隔物的 ELK16多核苷 酸按 LipA基因在 N端的顺序合成出来,再将此段多核苷酸*** pET^30a(+) 质粒的 Ndel和 Xhol位点间, 形成 pET^30a(卜)- LipA- ELK16。
使 用 Tiangen 公 司 的 高 纯 度 质 粒 小 提 试 剂 盒 提 取 pET~30a(H LipA-ELKl 6质粒和 New England Biolab (NEB)公司的 pTWINl 质粒, 分别利用如下两套正向引物和反向引物, 按照常规方法进行 PCR扩 增获得 LipA多核苷酸片段和 Mxe GyrA内含駄多核 酸片段:
第一套引物: 上游引物 5'- GCGATACATATGCACCATCACCATCA- 3'
(SEQ ID NO:9, 带下划线誠基为限制性内切酶 Nde I识别位点)和下游引物
ID NO: 10;
第 二 套 引 物 : 上 游 引 物
ID NO: 11 , 和 下 游 弓 i 物
5'-ATTTi: AAGCTTAGCGTGGCTGACGAACCCGTTC-3! (SEQ ID NO: 12, 带下划线碱基为限制性内切酶 Hind III识别位点)。
PCR反应使用 Tiangen公司的 pfo聚合酶, PCR反应条件为: 先 94°C 2min; 然后 94°C Imin, 57°C lmin, 72 °C 40sec, 共 30个循环; 最后 72 °C lOmiiio 反应结束后, 对 PCR扩增产物进行 1%琼)!糖凝胶电泳检测, 结果 PCR扩增出正确大小的条带, 与预期结果相符。 之后将两个片段进行 凝胶分离回收, 再以两个片段作为模板, 进行重叠 PCR反应: 先在不加入 引物的情况下 94 °C 2min; 然后 94 °C Imin, 70 °C Imiii, 72 °C 80sec, 共 10 个循环; 最后 72 °C 10min。 再进行 94 °C 2min ; 然后加入引物 5'-GCGATACATATGCACCATCACCATCA~3'(SEQ ID NO: 13) 和 5LATTTTAAAGCTTAGCGTGGCTGACGAACCCGTTC-3'(SEQ ID NO: 14), PCR程序 94 °C Imin, 57 °C Imin, 72 °C 40sec, 共 17个循环; 最后 72°C 10min。 反应结束后, 对 PCR扩增产物进行 1 琼脂糖凝胶电泳检测, 结果 PCR扩增出与预期相符的正确条带。 将重叠 PCR产物用限制性内切醸 Nde I 和 Hind III 进行双酶切 后 与 经 同样酶双酶切 的 质粒 pET-30a(+)-LipA-EI,K16 进行连接, 将连接产物转化到大肠杆菌 BL21(DE3XNovageii)感受态细胞, 将转化细胞涂布于添加有 50 g/mL卡那 霉素的 LB平板上筛选阳性克隆, 提取质粒, 对其进行测序, 测序结果表明 所克隆的 pE iipA- intein- ELK16序列正确。
再构建 pET rx-EK- Intein- ELK16载体, 该载体结构如图 ID所示。 先用表 1 中引物 Trx- For和 ΊΥχ- Rev, 以大肠 F菌菌株 BL21(DE3)的基 因组 UNA为模板, 扩增编码 Trx蛋白的基因 trxA, 用 Nde I和 Spe I限制 性内切酶双酶切后***同样处理后的 pET ipA- Intein- ELK16 载体进行连 接, 将连接产物转化到大肠杆菌 BL21(DE3)感受态细胞, 将转化细胞涂布 于添加有 50 g/mL卡那霉素的 LB平板上筛选阳性克隆, 提取质粒, 对其 进行测序, 测序结果表明获得正确的 pE Trx- Intein- ELK16。
之 后 用 表 1 中 弓 i 物 Trx- EK- For 和 Trx- EK-Rev , 以 pE LipA- Intein- ELK16为模板, 扩增带 EK位点的 intein基因, 并用 Bgl II 和 Hind III限制性内切酶双酶切后***同样处理后的 pET-Trx- intein- ELK16 质粒载体进行连接, 将连接产物转化到大肠杼菌 BL2i(DE3)感受态细胞, 将转化细胞涂布于添加有 50 μ8/πι£卡那霉素的 LB平板上筛选阳性克隆, 提取质粒,对其进行测序,测序结果表明所克隆的 pET-Trx-EK- intein- ELK16 序列正确。 表 1
引物 碱基序列 SEQ ID NO 说明
Trx- For 5'-AGTTACATATGAGCGATAAAATTATTC-3' 15 Nde I
5'-TCACGACTAGTGCATCTCCCGTGATGCACATTCGC 16 Spe I,
Trx-Rev ATGATATCAGAACCTGAACCTGAACCAGATCTCGCC EcoR V,
AGGTTAGCGTCGAGGAAC-3' Bgl 11
5'-AGTCAAGATCTGGGTACCGACGACGACGACAAGG 17
Trx-EK-For Bgl 11
ATATCATGCGAATGTGCATCACGGGAGATGC-3'
Trx-EK-Rev 5 '-GTCGGAAGCTTAGCGTGGCTGACGAACCCGTTC-3 ' 18 Hind III 实施例 2: 构建七种医药用多 jft的 ELK16融合表达构建体
选定了姨高血糖素样肽 GLP- 1、 B型脑利钠駄 BNP、 促姨岛素分泌 jft Ex- 4、 趋化因子 CCL5、 基质细胞衍生因子 SDF- ia、 促生长因子 IGF- 1α和 肥牌荷尔蒙 Lep作为目的多駄通过本发明的方法进行重组产生和纯化。 所 述多肽信息见下表 2: 表 2
目标 在大肠杆菌中的表达 氨基酸序列 优化的核苷酸序列 长度
多肽 (SEQ ID NO) (SEQ ID NO)
19 20
GLP- 1 31 aa 作为融合蛋白可溶, 否则将降解 (Wu等,
201 Ϊ )
作为融合蛋白可溶, 否则将降解 (Sim等, 2ί 22
BNP 32 aa
2005)
作为融合蛋白可溶, 否 ^将降解 23 24
Ex-4 39 aa
(Bosse-Doenecke等, 2008:)
CCL-5 66 aa 包涵体 (Proudfoot等, 1995) 25 26
SDF- ! a 67 aa 包涵体 (Cho等, 2008) 28
IGF- l a 70 aa 包涵体 (Zhang等, 2010) 29 30
Lep 346 aa 包涵体 (Gertler等, 1998) 31 32
对编码以上目的多肽的核苷酸序列进行密码子优化 (见表 2), 以便其能 更好地在大肠杆菌中表达, 并采用化学合成法获得相应编码序列。
下面以 GLP- 1为例说明七种医药用多肽的 ELK16融合表达构建体的构 建方法。
扩增 GLP 1所用的引物序列如下表 3所示。利用表 3中的引物 GLP1 F 和 GLP1 R, 通过 PCR扩增编码 GLP- 1的编码多核苷酸, 并在用 Nde I和 Spe I双酶切后将其***如图 1 C所示的表达载体 pET- LipA Intdn- ELK16同 样双醸切后的 Nde I与内含肽 (Intein)编码序列内部靠近 5'端的 Spe I位点之 间 (距离 5'端 17个碱基的位置), 同 保持内含肽 (Intein)编码序列 5'端的序 列不变; 另一方面, 利用表 3中的引物 Trx- GLP1- F和 GLP1- R扩增目的多 台, 并将其***双酶切后 pET-Trx- EK- Intein- ELK16的 Bgl II与 Spe I位点 之间, 同时在 Bgl II 位点与目的多肽之间引入肠激酶 (EK)切割位点 DDDDK(Asp- Asp- Asp- Asp- Lys)。
引物 碱基序列 SEQ ID NO 说明
GLP 1 -F S'-AGCATCATATGCATGCAGAAGGCACCTTT-B' 33 Nde I
S'-AGTCAAGATCTGGGTACCGACGACGACGACA 34 Trx- GLPl -F Bgl II
AG CATGCAGAAGGCACCTTT-3'
S'-AGCATACTAGTGCATCTCCCGTGATGCAGATAT 35
GLP1 - Sep I
CACCACGACCTTTAACCAG-3' 实施例 3 : 七种医药用多駄的表达和初步纯化
通过氯化钙法化将实施例 2 中获得的融合表达构建体分别转化入大肠 杆菌 BL21 (DE3)感受态细胞,通过菌落 PCR和质粒测序鉴定得到阳性克隆。
将阳性克隆接入 LB培养基中, 用 0.2 mM IPTG分别在 23 °C、 30°C、 37°C诱导表达 6 ho收获菌体,并测量菌浓度 OD6() (以下将 1 mL的 OD6«。 为 1的细胞量称为 1 OD)。
将菌体用裂解缓冲液 (2.4 g 的 Tris、 29.22 g 的 NaCl、 0,37 g 的 Na2 EDTA-2H20溶解于 800 mL水中, 调 pH至 8.2, 加水定容至 1 L)重悬至 20 OD/mL, 超声破碎。 在 4°C, 10000 rpm的条件 T离心 i0 min, 分别收集上 清和沉淀部分。 将沉淀部分用添加了 0.5 %的表面活性剂 Triton X- 100的裂 解缓冲液洗涤 i次 (去除细胞膜碎片等杂质), 再用裂解缓冲液洗涤 2次, 使 得 Triton X- 100基本去除。
将洗涤后的沉淀部分用含有 40 mM DTT的内含駄切割缓冲液 (0.62 g二 硫苏糖醇溶解于 100 mL裂解缓冲液中,置于- 20°C待用)充分重悬,每 20 OD 细胞破碎后的裂解沉淀部分 ffi 1 mL内含肽切割缓冲液重悬 (20 OD/mL) ,在 4 Ό下放置 24 h, 使得内含牍充分进行自切割。
最后, 通过离心分离上清和沉淀部分, 沉淀部分用与上一重悬步骤相 同的体积的裂解缓冲液重悬。
将通过以上操作所获得的聚集钵、 内含肽介导的自切割后的沉淀以及 内含駄介导的自切割后释放的上清用 4- 12% Bis Tris SDS-PAGE 或 12% Tris-Glysine SDS- PAGE分析其蛋白成分, 结果如图 2所示。 其中图 2A为 不带 T«标签的目的多肽的表达与纯化结果; 图 2B为带 T c标签的目的多 駄的表达与纯化结果。 泳道 a: 菌体破碎后沉淀部分; 泳道 b : 融合蛋白经 过内含駄介导的自切割并离心后沉淀部分; 泳道 c : 融合蛋白经过内含肽介 导的自切割后的离心后上清部分; 泳道 p : 经 HPLC精制纯化后的样品; 泳 道 1-3 :蛋白定量标准品 (Std),其中较大的条带为牛血清白蛋白 BSA(67 kD) , 上样量依次为 3 μ8、 1.5 μ , 0,75 tag;其中较小的条带为抑菌肽 Aprotinin(6,5 kD) , 上样量依次为〗,5 g、 0.75 μ8, 0,3 μ8 ; 泳道 4- 7 : 只含有牛血清蛋白 BSA的蛋白定量标准品, 上样量依次为 6 μ8、 3 μ8、 1 .5 和 0.75 g。 融 合蛋白、 切割后的 Intein- ELK16 以及目的多) ¾的位置由图例标出。 蛋白分 子量标准 Ml (14〜94 kD)和 M2(3.3〜20.1 kD)各个条带对应的分子量列于蛋白 胶图片左侧和右侧。
依照蛋白定量标准品, 应用 Bio- Rad公司的 Quantity ONE凝胶定量分 析软件对 标条带进行光密度分析, 可什算得出融合蛋白形成的聚集体产 量、 在内含肽介导的自切割之后释放到上清中的目的多肽产量、 以及 的 多駄在上清中的纯度, 结果如表 4所示。
Figure imgf000018_0001
a蛋白聚集体产量和 b内含肽介导的自切割后的目的多 ift产量 (以在菌浓度 006∞为 2时, 每升 LB培养基中的大肠杆菌细胞产生 2.66 mg细胞湿重 ΐί·算); c 内含肽介导的自切割效率 = 00%><(切割 前聚集体表达量—切割后聚集体剰余量 )/切割前聚集体产量; d回收率 =100%χ目的多肽实际产量 /蛋白 聚集体在完全切割的情况下 ¾生产目的多肽的理论产量; e 目的多駄在内含駄介导的自切割后位于 不可溶组分中。 结果
对于未添加 Trx标签的多肽:
(1) 七类中长多肽有五种目的多肽, 分别为 GLP1、 CCL5、 SDF- 1α、 IGF- 1 与 Lep, 可在与 Intein ELK16融合表达后形成大量聚集体, 表达量 为 28.4-44.6 g/mg细胞湿重。 此外, 目的多肽 BNP和 Ex 4在图 2中未给 出结果, 原因是在菌体破碎后的上清和沉淀中均未见外源蛋白表达。 (2) 目的多肽 GLP- 1可在内含肽介导的切割后直接释放到上清中,回收 率约为 46.8 %,产量为 1 .8 g/mg 细胞湿重。因此,上清中的目的多駄 GLP- 1 直接使用反相 HPLC精制纯化, 如图 2所示, 终产物纯度达 95%以上, 终 产量约为 0.8 g/mg细胞湿重。
(3) 虽然 IGFl o SDF- 1 α、 CCL- 5和 Leptin与 lutein- ELK16融合表达 大部分分布在聚集体中, 但经内含駄介导的自切割后在不可溶沉淀中。 对于与 Trx融合表达的目的多肽:
(1)六种目标駄段: GLP-1 , Ex- 4、 BNP、 CCL5、 SDF- 1α与 IGF- 1α与 Trx 标签及 lutein- ELKi 6 融合表达后均可大量形成聚集体, 表达量为 11.4-57,8 g/mg细胞湿重; 且在内含肽介导的自切割之后可释放到上清之 中, 产量为 3.4~13.4 g/mg细胞湿重, 纯度为 63.0%~79 7%。 目的多肽 Lep 在图中未给出结果, 原因是与 Trx标签融合后的 Lep在内含駄介导的自切 割后仍停留在不 溶沉淀中, 可考虑更换促溶肽标签进行 Lep的表达纯化。
(2) 在对可溶性的改进方面, 其中 GLP- i、 BNP、 Ex4和 IGF- l ot在内 含肽介导的自切割之后释放到上清中的比例较高, 回收率为 46 2~73.1%。 然而, CCL5和 SDF- l ot经内含肽介导的自切割之后释放到上清中的比例较 低, 使得肽段的回收率分别降至 26.5%和 19.3%。也可考虑更换促溶肽标签 提高这两种多肽经内含牍介导的自切割之后在上清中的比例。 实施例 4: 通过肠激酶切割进一步分离纯化目的多肽
对实施例 3中获得的与 Trx融合表达的目的多肽 GLP-1、 BNP、 Ex- 4、 CCL5、 SDF- 1 a与 IGF- 1 α进行进一步的分离纯化。
首先, 使用肠激酶 (Enterokinase, New England BioLabs, 货号 P8070S) 对上述几种经内含肽介导的自切割之后能释放到上清中的融合蛋白 Trx-駄 进行切割, 具体的操作步骤为:
使用截留分子量为 3K 的超滤器通过超滤将内含肽介导的自切割后的 上清中的内含駄切割缓冲液置换为肠激酶切割缓冲液 (20 mM Tris-IICl, 50 mM aCl, 2 mM CaC12 , ρΐϊ 8.0); 在含有目标融合蛋白的溶液中加入酶量 为 0.001 % (w/w)的肠激酶 (例如, 对于 1 mL目标融合蛋白含量为 1 mg/mL 的溶液, 加入 1 0 ng肠激酶), 在 23 °C恒温反应 1 6 h; 将经肠激酶切割所得 样品保存在- 70Ό条件下等待后续纯化操作。
接下来, 对于肠激酶切割后生成的 Trx标签与目的多肽, 可进一步通 过反相 HPLC对其进行纯化, 详细过程如下:
配置流动相 100% ¾0(含 0.1% TFA), 与流动相 B: 80% 乙腈 (含 0.12% TFA);在流速为 1 mL/min的条件下,使用含 5% B的流动相平衡 HPLC 色谱柱, 所使用的为反相 C18色谱柱 Diamonsif, 货号 99603); 采用梯度 洗脱, 使在 0~60 min内流动相组成由 5%上升至 80% B, 同时检测波长为 215 nm ¾键特征吸收波长>与 280 nm(氨基酸 1>τ、 Ti 与 Phe的共轭双键吸 收波长)的吸光度曲线; 使用高效液相色谱***自动收集器采集洗脱过程中 出现的吸收峰对应级分; 将吸收峰对应级分分装、 冻干并保存于 - 20°C条件 下, 水将冻干样品重悬后 j¾ SDS-PAGE检测吸收峰对应的级分, 并进一 步进行质谱检测。 以对 Trx- GLPi进行肠激酶切割后的的样品为例, 其通过 反相 HPLC分离的吸收峰图谱如图 3所示。
将通过以上操作所获得的经肠激酶切割前后的样品以及经反相 HPLC 纯化后的产物用 4-12% Bis- Tris SDS- PAGE分析其蛋白成分, 结果如图 4所 示。 其中泳道 c: 肠激酶切割前样品; 泳道 d: 肠激酶切割后的样品; 泳道 Trx-CCL5经肠激酶切割后的沉淀; 泳道 p; 经反相 HPLC精制纯化后 样品;泳道 1- 3 :蛋白定量标准品 (Std),其中较大的条带为牛血清蛋白 BSA(67 kD) , 上样量依次为 3 tig, 1。5 μ8、 0.75 §; 其中较小的条带为抑菌牍 Aprotinm(6.5 kD), 上样量依次为 1.5 μ§、 0.75 0.3 go 黑色箭头指示肠 激酶切割后产生的 ΤΓ 标签, 蓝色箭头指示肠激酶切割后产生的目的多肽。 蛋白分子量标准 Ml(14〜66 kD)和 M2(3,3〜20.1 kD)各个条带对应的分子量分 别列于蛋白胶图片左侧和右侧。
依照蛋白定量标准品, 应用 Bio Rad Quantity ONE对目标条带进行光 密度分析, 进一步计算得出样品中的目的多肽产量以及纯度。 结果
(1) 在以上切割条件下, 肠激酶对于 Trx- GLP1、 Trx- BNP、 Trx- Ex4、 Trx- SDF- la以及 Trx- CCL5的切割效率接近 100%, i均产生了条带位置正 确的 TlX 标签与 的多駄, 证明对于以上目的多肽, 使用肠激酶可以高效 地特异性切割。 (2) 对于结果中未显示的 TYx- IGF 1οι,经检测发现切割后的条带位置不 正确, 且推测为在 IGF- 1α序列中发生了 ^特异性切割, 因此 IGF- 1 α不适 合通过肠激酶切割进行分离纯化。
(3) 经肠激醸切割去除 Trx标签后, CCL5样品中出现大量不可溶沉淀, 将离心后的上清与沉淀经 SDS- PAGE检测, 发现 CCL5完全分布于不可溶 组分中, 而 Ι χ标签完全分布于可溶组分中。说明 CCL5在与 Trx融合表达 后虽然可溶性得到了提高, 但是并未完全正确折叠, 故在去除促溶肽标签 Trx后形成沉淀。 可考虑更换促溶駄标签进行 CCL5的表达纯化。
(4) 去除 Trx标签的 GLP1、 BNP、 Ex4与 SDF- lot可经反相 HPLC进行 有效的回收和纯化, 终产物纯度达 95%以上, 终产量约为 0.3- L8 g/mg细 胞湿重, 具体结果参见下表 5 :
Figure imgf000021_0001
3经过 HPLC精制纯化后的目的多駄的产量 (每升 LB培养基中的大涵杆菌细胞产生 2.66士 0.99 mg细胞湿 重 i†'算, 采用 Pierce® bicinchoninic acid (BCA) 试剂盒进行定量)。 b 括号中列出了经 HPLC纯化之后的最 终回收率, 由目的多肽的终产量与表 3中聚集体表达量之比计算得到 上述实施例的结果表明, 促溶肽标签可与自聚集肽组合进行多肽产生 和纯化, 即使给目的多肽加入促溶肽标签, 自聚集肽仍能介导融合蛋白形 成不溶聚集体, 适于通过离心沉淀或过滤等方法进行快速分离纯化。 并且 加入促溶肽标签 以改善目的多肽的表达以及提高目的多肽的溶解性。 实施例 5: 利用不同促溶駄标签和自聚集駄的组合来产生和纯化多肽 促溶肽标签
除了 Trx外, 还选择了包括下表中的另外 5种常用促溶牍标签用于与 自聚集) ¾组合进行多肽产生和纯化。
Figure imgf000022_0001
根据上面实施例中描述的方法, 分别构建了融合蛋白 SUMO-胸腺素 (Thymosm)/BNP-intein-ELKl 6 、 GST-GLP 1 -intein-ELKl 6 、 DsbC- GLPi- intein- ELKi6、 GBi- GLPi- intein- ELK16、 Z-GLP1 -intein-ELKl 6 的表达载体, 并根据上面描述的方法进行了重组表达和基于自聚集肽的分 离纯化。 SDS- PAGE检测结果示于图 5- 9, 其中泳道 S: 细胞裂解物的可溶 部分; 泳道 ir 细胞裂解物的不可溶部分; 泳道 c5s; 经切割的融合蛋白的 可溶部分; 泳道 c n: 经切割的融合蛋白的不可溶部分。表达和纯化结果总 结于下表 7。
Figure imgf000022_0002
结果表明, 多种促溶牍标签均 与自聚集肽组合进行多肽产生和纯化。 自聚集) ¾ 18A和 L6KD
还对另外两种与 ELK16具育类似功能的自聚集肽〗 8A和 L6KD进行了 测试。 根据上面实施例中描述的方法, 分别构建了融合蛋白 Trx- GLPl- intein- 18A、 Trx- GLPl- intein- L6KD的表达载体, 并根据上面描述 的方法进行了重组表达和基于自聚集肽的分离纯化。 SDS- PAGE 检测结果 示于图 〗0, 其中泳道 s: 细胞裂解物的可溶部分; 泳道 in: 细胞裂解物的 不可溶部分; 泳道 c,s: 经切割的融合蛋白的可溶部分; 泳道 Cjf 经切割 的融合蛋白的不可溶部分。 结果表明自聚集駄 18A和 L6KD同样适用于本 发明的方法。 参考文献
Leader B, et al. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov, 2008, 7(l):21-39.
Bray B L. Large-scale manufacture of peptide therapeutics by chemical synthesis. Nat Rev Drug
Discov, 2003, 2(7):587-593.
Kamionka M. Engineering of therapeutic proteins production in Escherichia coli. Curr Pharm Biotechnol, 2011, 12(2):268-274.
Demain A L, Vaishnav P. Production of recombinant proteins by microbes and higher organisms. Biotechnol Adv, 2009, 27(3):297-306.
Walsh G Biopharmaceutical benchmarks 2006. Nat Biotechnol, 2006, 24(7):769-776.
Walsh G Biopharmaceutical benchmarks 2003. Nat Biotechnol, 2003, 21(8):865-870.
Kuliopulos A, Walsh C T. Production, purification, and cleavage of tandem repeats of recombinant peptides. J Am Chem Soc, 1994, 116(l l):4599-4607.
Hannig G, Makrides S C. Strategies for optimizing heterologous protein expression in Escherichia coli.
Trends Biotechnol, 1998, 16(2):54-60.
Murby M, et al. Upstream strategies to minimize proteolytic degradation upon recombinant production in Escherichia coli. Protein Expr Purif, 1996, 7(2): 129-136.
陈浩等人, 中国生物工程杂志, 2002, 22(5):p. 87-92.
Zhao X B, et al. Molecular self-assembly and applications of designer peptide amphiphiles. Chem Soc
Rev, 2010, 39: 3480-3498.
Mitraki A. Protein aggregation: from inclusion bodies to amyloid and biomaterials, in Advances in Protein Chemistry and Structural Biology. Elsevier Academic Press Inc: San Diego, 2010, 79: 89-125.
Wu X Y, et al. EW, a novel recombinant analogue of exendin-4 expressed in Escherichia coli. Scientific Research and Essays Vol. 6(14), pp. 2941-2949, 18 July, 2011.
Sun Z Y, et al. Use of Ssp dnaB derived mini-intein as a fusion partner for production of recombinant human brain natriuretic peptide in Escherichia coli. Protein Expression and Purification 43 (2005) 26-32.
Bosse-Doenecke, et al. High yield production of recombinant native and modified peptides exemplified by ligands for G-protein coupled receptors. Protein Expression and Purification 58 (2008) 114-121.
Proudfoot, et al. Extension of Recombinant Human RANTES by the Retention of the Initiating Methionine Produces a Potent Antagonist. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 271, No. 5, Issue of February 2, pp. 2599-2603, 1996
Cho, et al. Maltose binding protein facilitates high-level expression and functional purification of the chemokines RANTES and SDF-la from Escherichia coli. Protein Expression and Purification 60 (2008) 37-45
Zhang, et al. High-level soluble expression of hIGF-1 fusion protein in recombinant Escherichia coli.
Process Biochemistry 45 (2010) 1401-1405
Gertler, et al. Large-scale preparation of biologically active recombinant ovine obese protein (leptin). FEBS Letters 422 (1998) 137-140.
Leder et al. New methods for efficient protein production in drug discovery. Current Opinion in Drug Discovery & Development 2007 10(2): 193-202.
Esposito and Chatterjee Enhancement of soluble protein expression through the use of fusion tags. Current Opinion in Biotechnology 2006, 17:353-358.
Waugh, Making the most of affinity tags. TRENDS in Biotechnology Vol.23 No.6 June 2005.
Wu et al. Protein trans -splicing and functional mini-inteins of a cyanobacterial dnaB intein. Biochimica et Biophysica Acta 1387 (1998) 422-432.
TELENTI et al. The Mycobacterium xenopi GyrA Protein Splicing Element: Characterization of a Minimal Intein. JOURNAL OF BACTERIOLOGY, Oct. 1997, p. 6378-6382.
Zhang et al. Spontaneous assembly of a self-complementary oligopeptide to form a stable microscopic membrane. Proc. Natl. Acad. Sci. USA (1993) Vol 90, pp. 3334-3338.

Claims

1. 融合蛋白, 其包含促溶) ¾标签部分、 自聚集) ¾部分以及目的多肽部 分, 其中所述目的多駄部分位于所述促溶肽标签部分和所述自聚集肽部分 之间, 并且所述目的多肽部分通过第一接头连接于所述自聚集肽部分, 其 中所述第一接头包含第一切割位点, 所述融合蛋白在宿主细胞内表达后可 又
通过所述自聚集肽部分形成活性聚集体。
2. 权利要求 1的融合蛋白, 其中所述自聚集肽部分包含两亲性自组装 短肽。
3. 权利要求 2的融合蛋白, 其中所述两亲性自组装短牍选自两亲性 β 折叠短肽、 两亲性 α螺旋短肽和类表面活性剂短肽。
4. 权利要求 3的融合蛋白, 其中所述自聚集肽部分包括 1个所述两亲 性 β折叠短肽。 书
5. 权利要求 3的融合蛋白, 其中所述自聚集肽部分包括串联重复的两 个或更多个所述两亲性 β折叠短肽。
6. 权利要求 4或 5的融合蛋白, 其中所述两亲性 β折叠短肽的长度为 4- 30个氨基酸残基。
7. 权利要求 4-6任一项的融合蛋白, 其中所述两亲性 β折叠短肽的疏 水性氨基酸残基含量为 40% 80%。
8. 权利要求 7的融合蛋白, 其中所述两亲性 β折叠短肽的氨基酸序列 示于 SEQ ID ΝΟ: 1。
9. 权利要求 3的融合蛋白, 其中所述自聚集肽部分包括 1个所述两亲 性 α螺旋短肽。
10. 权利要求 3的融合蛋白,其中所述自聚集肽部分包括串联重复的两 个或更多个所述两亲性 α螺旋短肽。
11. 权利要求 9或 10的融合蛋白, 其中所述两亲性 α螺旋短肽的长度 为 4-30个氨基酸残基。
12. 权利要求 941任一项的融合蛋白, 其中所述两亲性 α螺旋短肽的 疏水性氨基酸残基含量为 40%- 80%。
13. 权利要求 12的融合蛋白, 其中所述两亲性 α螺旋短肽的氨基酸序 列示于 SEQ ID NO:2 o
14. 权利要求 3的融合蛋白, 其中所述类表面活性剂短肽具有 7-30个 氨基酸残基, 其从 N端到 C端具有以下通式表示的氨基酸序列:
A-B或 B-A
其中 A是由亲水性氨基酸残基组成的肽, 所述亲水性氨基酸残基可以 是相同的或不同的, 且选自 Lys、 Asp. Arg、 Glu、 His. Ser、 Thr、 Asn和 Gin;
B 是由疏水性氨基酸残基组成的肽, 所述疏水性氨基酸残基可以是相 同的或不同的, 且选自 Leu、 Gly、 Ala. Val、 Ile、 Phe和 Tip;
A与 B通过肽键连接; 并且
其中在所述类表面活性剂短肽中疏水性氨基酸残基的比例是 55%-95%。
15. 权利要求 14的融合蛋白, 其中所述类表面活性剂短肽具有 8个氨 基酸残基, 其中在所述类表面活性剂短肽中疏水性氨基酸残基的比例是 75%。
16. 权利要求 15的融合蛋白, 其中所述类表面活性剂短肽的氨基酸序 列示于 SEQ ID NO:3、 SEQ ID NO:4或 SEQ ID NO:5。
17. 权利要求 146中任一项的融合蛋白, 其中所述自聚集肽部分位于 所述融合蛋白的 C端。
18. 权利要求 1-17 中任一项的融合蛋白, 其中所述促溶肽标签选自 NusA、 GST, Trx、 SUMO, DsbC、 Z八 GB1 、 MBP禾 Π T7PK:。
19. 权利要求 148 中任一项的融合蛋白, 其中所述第一切割位点选自 化学切割位点、 醇法切割位点和自切割位点。
20. 权利要求 19的融合蛋白, 其中所述自切割位点为内含肽。
21. 权利要求 20的融合蛋白,其中所述内含肽为序列示于 SEQ ID NO:6 的 Mxe GyrA
22. 权利要求 21的融合蛋白, 其中所述 Mxe GyrA直接连接于所述目 的多肽的 C端。
23. 权利要求 1-22 中任一项的融合蛋白, 其中所述第一接头还包含间
24. 权利要求 1-23 中任一项的融合蛋白, 其中所述 的多肽通过第
25. 权利要求 24的融合蛋白, 其中所述第二接头包含间隔物。
26. 权利要求 24的融合蛋白, 其中所述第二接头包含第二切割位点, 其中第二切割位点的切割条件不同于所述第一切割位点的切割条件。
27. 权利要求 26的融合蛋白, 所述第二切割位点选自第二化学切割位 点、 第二酶法切割位点和第二自切割位点。
28. 权利要求 27的融合蛋白,其中所述第二酶法切割位点包含 SEQ ID NO:7所示的肠激酶识别序列。
29. 权利要求 28的融合蛋白, 其中所述肠激酶识别序列直接连接于所 述目的多肽的 N端。
30. 权利要求 26- 29中任一项的融合蛋白, 其中所述第二接头还包括间
31. 权利要求 1-30中任一项的融合蛋白, 其中所述目的多肽为长度为 30-100个氨基酸残基的多肽。
32. 多核苷酸, 其包含编码权利要求 1-31 中任一项的融合蛋白的核苷 酸序列或其互补序列。
33. 表达构建体, 其包含权利要求 32的多核苷酸。
34. 宿主细胞,其包含权利要求 32的多核苷酸或以权利要求 33的表达 构建体转化, 其中所述宿主细胞能够表达所述融合蛋白。
35. 产生和纯化目的多肽的方法, 所述方法包括以下步骤:
(a) 培养权利要求 34的宿主细胞, 从而表达所述融合蛋白;
(b) 裂解所述宿主细胞, 然后去除细胞裂解物的可溶部分, 回收不溶部 分;
(c) 通过切割第一切割位点 所述不溶部分释放可溶的带有促溶馱标 签的目的多肽; 和
(d) 去除步骤 (c)中的不溶部分, 回收含有所述目的多肽的可溶部分。
36. 权利要求 35的方法, 还包括:
(e) 如果存在第二切割位点,则通过切割第二切割位点使所述目的多牍 和所述促溶肽标签分离;
(f 去除所述促溶肽标签, 获得纯化的目的多駄。
PCT/CN2012/082879 2012-10-12 2012-10-12 多肽的产生和纯化方法 WO2014056199A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
PCT/CN2012/082879 WO2014056199A1 (zh) 2012-10-12 2012-10-12 多肽的产生和纯化方法
CN201280076298.3A CN104755502B (zh) 2012-10-12 2012-10-12 多肽的产生和纯化方法
US13/836,817 US9200306B2 (en) 2012-10-12 2013-03-15 Methods for production and purification of polypeptides

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/082879 WO2014056199A1 (zh) 2012-10-12 2012-10-12 多肽的产生和纯化方法

Publications (1)

Publication Number Publication Date
WO2014056199A1 true WO2014056199A1 (zh) 2014-04-17

Family

ID=50475660

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/082879 WO2014056199A1 (zh) 2012-10-12 2012-10-12 多肽的产生和纯化方法

Country Status (3)

Country Link
US (1) US9200306B2 (zh)
CN (1) CN104755502B (zh)
WO (1) WO2014056199A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104263747A (zh) * 2014-09-22 2015-01-07 程永升 一种长效蛋白质药物表达复性修饰的双标签载体及方法
WO2021083301A1 (zh) * 2019-10-31 2021-05-06 华南理工大学 多肽的生产和纯化方法
CN113637698A (zh) * 2021-07-27 2021-11-12 武汉菲恩生物科技有限公司 一种串联双促溶标签及其应用
WO2024092879A1 (zh) * 2022-10-31 2024-05-10 杭州先为达生物科技股份有限公司 高特异性肠激酶酶切方法及乙醇在提高肠激酶酶切特异性中的用途

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016065273A1 (en) * 2014-10-24 2016-04-28 The University Of Chicago Heat-inducible self-assembling protein domains
CN107075491B (zh) 2014-10-28 2021-07-06 谷万达公司 用于稳定反式剪接的内含肽修饰的蛋白酶的方法和组合物
US20180340156A1 (en) * 2015-09-10 2018-11-29 The University Of Toledo Purification of a soluble and active form of aspartate n-acetyltransferase
CN106986922B (zh) * 2017-04-14 2020-03-06 江南大学 一种自组装双亲短肽及其应用
CN108531492A (zh) * 2018-04-23 2018-09-14 内蒙古农业大学 利用类弹性蛋白elp构建活性绵羊β-防御素sbd-1体外表达的方法
WO2020048996A1 (en) 2018-09-06 2020-03-12 ETH Zürich Self-assembling globular proteins and uses thereof
CN109627290B (zh) * 2018-12-12 2022-03-29 华南理工大学 α螺旋自组装短肽及其在蛋白质纯化中的应用
CN113195521B (zh) * 2018-12-19 2023-05-12 清华大学 Mtu ΔI-CM内含肽变体和其应用
US11208477B2 (en) 2019-04-01 2021-12-28 Novo Nordisk A/S Antibodies and use thereof
WO2020233685A1 (zh) * 2019-05-21 2020-11-26 广州市妇女儿童医疗中心 一种基于内含肽介导纳米载体及其应用与一种可同时递送抗原和免疫增强剂的纳米制剂
CN110272498A (zh) * 2019-05-21 2019-09-24 广州市妇女儿童医疗中心 一种基于内含肽介导纳米载体及其应用
CN110354271A (zh) * 2019-05-21 2019-10-22 广州市妇女儿童医疗中心 一种基于内含肽介导纳米载体模块平台及其构建方法与应用
CN110201183A (zh) * 2019-05-21 2019-09-06 广州市妇女儿童医疗中心 一种可同时递送抗原和免疫增强剂的纳米制剂
CN110484518B (zh) * 2019-07-03 2023-11-10 天津科技大学 一种自组装短肽标签标记的氟化酶聚集体及应用
GB201917046D0 (en) * 2019-11-22 2020-01-08 Ge Healthcare Bioprocess R&D Ab Improved protein production
CN114317577A (zh) * 2020-09-28 2022-04-12 江苏万邦医药科技有限公司 一种可溶性表达***及其在蛋白可溶性表达中的应用
WO2022253266A1 (zh) * 2021-06-03 2022-12-08 华南理工大学 重组蛋白纯化方法
EP4379054A1 (en) * 2021-07-27 2024-06-05 Stand Therapeutics Co., Ltd. Peptide tag and nucleic acid encoding same
WO2023108026A2 (en) 2021-12-08 2023-06-15 Oakgrove Bio Llc Biological production of histidine-rich peptides
CN114540392A (zh) * 2022-03-02 2022-05-27 南京市第一医院 一种碱性成纤维细胞生长因子制备方法及其应用
CN115896144B (zh) * 2022-10-17 2024-01-02 湖南诺合新生物科技有限公司 Fus蛋白在作为融合标签中的应用,重组蛋白及其表达方法

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060263855A1 (en) * 2005-03-14 2006-11-23 Wood David W Intein-mediated protein purification using in vivo expression of an elastin-like protein
CN102226172A (zh) * 2011-05-09 2011-10-26 清华大学 基于自聚集短肽诱导的酶聚集体的蛋白质纯化方法

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5821088A (en) * 1990-05-11 1998-10-13 Siga Pharmaceuticals, Inc. Use of gram-positive bacteria to express recombinant proteins
ATE298796T1 (de) * 1998-10-13 2005-07-15 Univ Georgia Res Found Stabilisierte bioaktive peptide und methoden zu ihrer identifikation, synthese und verwendung
US20020102703A1 (en) * 1999-12-29 2002-08-01 Sheppard Paul O. Kunitz domain polypeptide zkun10
WO2001083804A2 (en) * 2000-05-03 2001-11-08 Expressive Constructs, Inc. A method for improving recombinant protein stability and solubility
US20040142429A1 (en) * 2001-01-25 2004-07-22 Grant Chris W.M. Method for the expression, purification, and structure recovery and handling of hydrophobic proteins, polypeptides or peptides
DE60331601D1 (de) * 2003-12-18 2010-04-15 Univ Giessen Justus Liebig Neue chimärische plasminogenaktivatoren und deren pharmazeutische verwendung
EP2860194B1 (en) * 2006-09-26 2018-11-07 Massachusetts Institute Of Technology Modified self-assembling peptides
AU2007329171A1 (en) * 2006-12-04 2008-06-12 Innovative Purification Technologies Pty Ltd Protein particles
US7678883B2 (en) * 2007-07-25 2010-03-16 E.I. Du Pont De Nemours And Company Solubility tags for the expression and purification of bioactive peptides
ES2763168T3 (es) * 2009-06-03 2020-05-27 Basf Se Producción recombinante de péptidos
CA2792476C (en) * 2010-03-18 2018-12-11 Spiber Technologies Ab Production of proteins and polypeptides
US8940858B2 (en) * 2010-05-17 2015-01-27 Northwestern University Fibrous microcapsules and methods of assembly and use thereof
CN103732758A (zh) * 2010-12-21 2014-04-16 香港中文大学 表达和纯化植物中重组蛋白的低成本方法
US20120277410A1 (en) * 2011-03-30 2012-11-01 Molecular Kinetics Incorporated Dehydrin-based entropic bristle domain sequences and their use in recombinant protein production

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060263855A1 (en) * 2005-03-14 2006-11-23 Wood David W Intein-mediated protein purification using in vivo expression of an elastin-like protein
CN102226172A (zh) * 2011-05-09 2011-10-26 清华大学 基于自聚集短肽诱导的酶聚集体的蛋白质纯化方法

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GE XIN ET AL.: "Self-Cleavable Stimulus Responsive Tags for Protein Purification without Chromatography", J. AM. CHEM. SOC., vol. 127, 22 July 2005 (2005-07-22), pages 11228 - 11229 *
LI, YONGJIN ET AL.: "Fusion Tags Technology and Their Applications", JOURNAL OF BIOTECHNOLOGY, vol. 22, no. 4, 31 July 2006 (2006-07-31), pages 523 - 527 *
XING, LEI ET AL.: "Streamlined protein expression and purification using cleavable self-aggregating tags, abstract 42", MICROBIAL CELL FACTORIES, vol. 10, 2 June 2011 (2011-06-02), pages 1 - 7 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104263747A (zh) * 2014-09-22 2015-01-07 程永升 一种长效蛋白质药物表达复性修饰的双标签载体及方法
WO2021083301A1 (zh) * 2019-10-31 2021-05-06 华南理工大学 多肽的生产和纯化方法
CN113637698A (zh) * 2021-07-27 2021-11-12 武汉菲恩生物科技有限公司 一种串联双促溶标签及其应用
WO2024092879A1 (zh) * 2022-10-31 2024-05-10 杭州先为达生物科技股份有限公司 高特异性肠激酶酶切方法及乙醇在提高肠激酶酶切特异性中的用途

Also Published As

Publication number Publication date
CN104755502A (zh) 2015-07-01
US9200306B2 (en) 2015-12-01
US20140106399A1 (en) 2014-04-17
CN104755502B (zh) 2018-05-18

Similar Documents

Publication Publication Date Title
WO2014056199A1 (zh) 多肽的产生和纯化方法
US8796431B2 (en) Efficient production of peptides
WO2021083301A1 (zh) 多肽的生产和纯化方法
JPH074255B2 (ja) 形質転換宿主細胞中にハイブリッド・ポリペプチドを発現させるdna発現ベクター
WO2022241831A1 (zh) 多肽的制备方法
WO2014170430A1 (en) Methods for the expression of peptides and proteins
CN111132996A (zh) 用于重组蛋白表达的融合标签
US20110092424A1 (en) Production of glucagon like peptide 2 and analogs
JP4088584B2 (ja) 融合タンパク質から目的タンパク質を分離する方法。
WO2020187270A1 (zh) 含有荧光蛋白片段的融合蛋白及其用途
CN107629129B (zh) 生产和纯化多肽的方法
US20150315259A1 (en) Method of producing a recombinant peptide
WO2022208554A2 (en) Constructs and methods for increased expression of polypeptides
CN117801123B (zh) 沃索利肽可溶性中间体、中间体制备方法及沃索利肽的制备方法
KR20100020784A (ko) 시스토비루스 phi6의 외피막 단백질 P9을 융합파트너로포함하는 막 단백질 발현벡터 및 이를 이용한 막 단백질의생산방법
CN117802138B (zh) 利那洛肽可溶性中间体和利那洛肽的制备方法
CN114774397B (zh) 牛肠激酶轻链蛋白突变体及重组融合蛋白
CN114807101B (zh) 包含牛肠激酶轻链蛋白的融合蛋白及其表达载体和重组工程菌
WO2024119434A1 (zh) 一种提高重组蛋白表达效率的酸性表面助溶短肽标签
CN116496365B (zh) 一种提高重组蛋白表达效率的酸性表面助溶短肽标签
WO2003046158A1 (en) A method for preparing the recombinant human glucagon like peptide-1 (7-37)
CN116496365A (zh) 一种提高重组蛋白表达效率的酸性表面助溶短肽标签
JP2020513834A (ja) ペプチドの発現および大規模生産
KR20210079235A (ko) Whep 도메인 융합에 의한 목적 단백질의 수용성 증진 방법
BR112015026384B1 (pt) Método para produzir peptídeo recombinante (peoi) ou proteína de interesse (proi)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12886241

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12886241

Country of ref document: EP

Kind code of ref document: A1