CN109321493B - B-lysine-resistant bacillus ZJB-17007 and application thereof - Google Patents

B-lysine-resistant bacillus ZJB-17007 and application thereof Download PDF

Info

Publication number
CN109321493B
CN109321493B CN201811153717.4A CN201811153717A CN109321493B CN 109321493 B CN109321493 B CN 109321493B CN 201811153717 A CN201811153717 A CN 201811153717A CN 109321493 B CN109321493 B CN 109321493B
Authority
CN
China
Prior art keywords
phenylacetyl
reaction
solution
phosphoryl
methyl
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN201811153717.4A
Other languages
Chinese (zh)
Other versions
CN109321493A (en
Inventor
柳志强
郑裕国
康雪梅
张晓健
金利群
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zhejiang University of Technology ZJUT
Original Assignee
Zhejiang University of Technology ZJUT
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 Zhejiang University of Technology ZJUT filed Critical Zhejiang University of Technology ZJUT
Priority to CN201811153717.4A priority Critical patent/CN109321493B/en
Publication of CN109321493A publication Critical patent/CN109321493A/en
Application granted granted Critical
Publication of CN109321493B publication Critical patent/CN109321493B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • C12N1/20Bacteria; Culture media therefor
    • C12N1/205Bacterial isolates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12RINDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
    • C12R2001/00Microorganisms ; Processes using microorganisms
    • C12R2001/01Bacteria or Actinomycetales ; using bacteria or Actinomycetales
    • 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
    • C12N1/20Bacteria; Culture media therefor
    • 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
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/04Alpha- or beta- amino acids

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to a boron-resistant lysine bacillus (Lysinibacillus boronitolorans) ZJB-17007 and application thereof, wherein the selectivity value of L-amino acid enantiomer produced by catalyzing N-phenylacetyl-DL-amino acid reaches 99%, and the selectivity value of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid enantiomer prepared by catalyzing 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid reaches 99%.

Description

B-lysine-resistant bacillus ZJB-17007 and application thereof
(I) technical field
The invention relates to a bacterial strain for producing amidohydrolase, namely boron-resistant lysine bacillus (Lysinibacillus boroniculatus) ZJB-17007, and application thereof in catalyzing N-phenylacetyl amino acid to prepare chiral amino acid and catalyzing N-phenylacetyl substituent of amino acid derivative to prepare chiral amino acid derivative.
(II) background of the invention
The chemical name of the L-glufosinate-ammonium is as follows: 4- [ hydroxyl (methyl) phosphonyl ] -L-homoalanine is a structural analogue of L-glutamic acid, can inhibit the activity of Glutamine Synthetase (GS), enables ammonia accumulation in plants, blocks the light respiration of the plants due to high-concentration ammonia accumulation, destroys chloroplast structures and cysts of matrixes, and simultaneously inhibits amino acid synthesis to damage cell membranes and kill cells, thereby killing weeds. It has broad spectrum and is herbicide tolerant to the second major transgenic crop in the world.
The existing L-glufosinate-ammonium synthesis method comprises a chemical synthesis method and a biological synthesis method. The chemical method mainly constructs the chiral center of L-glufosinate-ammonium by the following 4 ways: 1) constructing a chiral center by chiral induction by using a chiral auxiliary reagent; 2) taking natural amino acid as a chiral source, and converting to obtain L-glufosinate-ammonium; 3) asymmetric synthesis reaction catalyzed by chiral catalyst; 4) chiral resolution of the racemate. However, the chemical method for synthesizing L-glufosinate-ammonium has the disadvantages of multiple working procedures, low yield, high chiral reagent cost, large amount of three wastes and difficult treatment. The biological method has the advantages of strict stereoselectivity, mild reaction conditions, high yield, easy separation and purification and the like, so that the technology for producing the L-glufosinate-ammonium by the biological method has very important industrial development value.
Biological production of L-glufosinate-ammonium according to the substrate, there are protease hydrolysis bialaphos, hydrolysis of L-3-acetamido-4- (hydroxymethylphosphono) butanamide by phosphodiesterase I, amidase I and glutaminase stepwise synergy maintaining optical activity, resolving bialaphos ethyl ester by alpha-chymotrypsin, deacetylase resolution of N-acetyl-glufosinate, amidase resolution of 2-amino-4- (hydroxymethylphosphono) -butanamide, ester hydrolase hydrolysis of L-glufosinate-N-carboxylic acid anhydride, nitrile hydratase hydrolysis of glufosinate nitrile-containing substrate, transaminase catalysis of 2-carbonyl-4- (hydroxymethylphosphono) butyric acid and the like.
The existing chemical method for synthesizing the L-glufosinate-ammonium mainly comprises the following steps: 1) the chiral auxiliary inducing method is characterized in that (S) -2-hydroxy-3-pinone is used as the chiral auxiliary for preparation, the yield is 51 percent, and the e.e. value is 79 percent; the D-valine methyl ester is used as a chiral auxiliary agent, the reaction is carried out at a low temperature of-78 ℃, the yield is 51 percent, and the e.e. value is 93.5 percent. 2) The natural amino acid chiral source method takes L glutamic acid as a chiral source, and the e.e. value is 99.4 percent; l-methionine is chiral source, total yield is 42.3%, e.e. value is 93.5%, and virulent iodomethane is needed. 3) Asymmetric synthesis method, asymmetric catalytic hydrogenation-large catalyst dosage and expensive trimethyl silicon cyanide price; asymmetric Strecker reaction-use highly toxic cyanide; asymmetric Michael addition-the catalyst consumption is large, and the product yield and the e.e. value are low. 4) The racemate resolution method has the highest yield of 86 percent and the highest e.e. value of 99 percent, but the D-glufosinate-ammonium is not converted after the resolution, so that the raw materials are wasted.
The important effective component 2-amino-4- [ hydroxyl (methyl) phosphoryl ] -L-butyric acid of the L-glufosinate-ammonium is prepared by selectively catalyzing and resolving 2-N-phenylacetyl-4- [ hydroxyl (methyl) phosphoryl ] -DL-butyric acid by using amidohydrolase, and a basis is provided for realizing the synthesis of the L-glufosinate-ammonium in an industrial route.
Chiral amino acids and derivatives thereof have an increasing role in pharmaceutical development, and the current pharmaceutical development has an increasing demand for chiral purity. The chiral amino acid production process includes chemical resolution, enzyme resolution and other processes. The enzymatic resolution has the advantages of mild condition, strong specificity, less pollution and obvious advantages compared with the chemical resolution. The amide hydrolase selectively catalyzes and resolves the N-phenylacetyl amino acid to produce the chiral amino acid, so that the chiral amino acid has wide application prospect.
Disclosure of the invention
The invention aims to provide a bacterial strain capable of producing amidohydrolase, namely, boron-resistant lysine bacillus (lysine bacillus boronitolorans) ZJB-17007, and application thereof in catalyzing N-phenylacetyl-DL-amino acid derivatives (2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid) to prepare L-glufosinate-ammonium important effective components, namely L-amino acid derivatives (2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid) or catalyzing N-phenylacetyl-DL-amino acid to prepare L-amino acid, so that a new enzyme source is provided for producing L-glufosinate-ammonium effective components and L-amino acid by a chemical-enzymatic method, and the development of green chemical catalysis is promoted.
The technical scheme adopted by the invention is as follows:
in a first aspect, the invention provides an amidohydrolase-producing strain, namely, a boron-resistant lysine bacillus (lysine bacillus boronolans) ZJB-17007, which is preserved in China center for type culture Collection with the preservation number of CCTCC No: m2017602, date of deposit 2017, 10 month 23 day, address: wuhan university, 430072. The strain is separated from soil by the institute of bioengineering of Zhejiang industrial university, and has the capability of catalyzing and synthesizing L-glufosinate-ammonium through detection.
In a second aspect, the present invention provides an application of said boron-tolerant lysinibacillus bornitolerans ZJB-17007 in the microbial catalytic resolution of N-phenylacetyl-DL-amino acid derivatives (preferably 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid) to the preparation of L-amino acid derivatives (preferably 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid), which are important active ingredients of L-glufosinate, said application being: using wet thalli obtained by fermentation culture of B-lysine-resistant bacillus ZJB-17007 as a catalyst, using N-phenylacetyl-DL-amino acid derivatives (preferably 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid) as a substrate, using a buffer solution (preferably ammonia water) as a reaction medium to form a reaction system with a pH value of 8.5, carrying out conversion reaction at 25-55 ℃, 100-200rpm (preferably 30-40 ℃, 150rpm), and after the reaction is finished, separating and purifying the reaction solution to obtain L-amino acid derivatives (preferably 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid). In the reaction system, the dosage of the catalyst is 10-200g/L, preferably 50g/L, and the final concentration of the substrate added initially is 10-500mM, preferably 100mM, calculated by the weight of wet bacteria. The reaction system can also be composed of fermentation liquor and substrate which are obtained by fermenting and culturing the B-resistant lysine bacillus ZJB-17007, and the pH value is 8.5; wherein the final concentration of wet thallus in the fermentation liquor in the whole reaction system is 10-200g/L, preferably 50g/L, and the final concentration of the substrate in the reaction system is 10-500mM, preferably 100 mM.
The method for separating and purifying the reaction liquid comprises the following steps: after completion of the reaction, the reaction mixture is extracted with methylene chloride, the pH of the aqueous layer is adjusted to 1.0 to 5.0 (preferably 2.5), loading the sample at a rate of 1-6.0Bv/h (preferably 4Bv/h) for ion exchange chromatography, washing with deionized water, eluting with 0.5-3.0Bv/h (preferably 2Bv/h) with 0.2-4.5M (preferably 1M) ammonia water, detecting the eluate with 0.2% ninhydrin solution-impregnated filter paper, collecting eluate containing target components, distilling under reduced pressure to paste, dissolving in methanol for recrystallization, and drying to obtain L-amino acid derivative (preferably 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid).
In a third aspect, the invention also provides an application of the boron-resistant lysine bacillus ZJB-17007 in catalyzing N-phenylacetyl-DL-amino acid to prepare L-amino acid, wherein wet thallus obtained by fermentation culture of the boron-resistant lysine bacillus ZJB-17007 is used as a catalyst, N-phenylacetyl-DL-amino acid is used as a substrate, a buffer solution (preferably ammonia water) is used as a reaction medium to form a conversion system with the pH of 8.5, the conversion reaction is carried out under the conditions of 25-55 ℃, 100-200rpm (preferably 30-40 ℃ and 150rpm), and after the reaction is finished, the reaction solution is separated and purified to obtain the L-amino acid. In the transformation system, the dosage of the catalyst is 20-300g/L (preferably 20-60g/L) based on the weight of wet bacteria, and the final concentration of the substrate added is 50-500mM (preferably 100 mM). The N-phenylacetyl-DL-amino acid is one of the following: N-phenylacetyl-DL-alanine, N-phenylacetyl-DL-serine, N-phenylacetyl-DL-glutamic acid, N-phenylacetyl-DL-tyrosine, N-phenylacetyl-DL-threonine, N-phenylacetyl-DL-valine, N-phenylacetyl-DL-aspartic acid, N-phenylacetyl-DL-methionine, N-phenylacetyl-DL-leucine, N-phenylacetyl-DL-isoleucine, N-phenylacetyl-DL-phenylalanine. The method for separating and purifying the reaction liquid comprises the following steps: after the reaction is finished, extracting the reaction solution by using dichloromethane, adjusting the pH of an aqueous layer to 1.0-5.0 (preferably 2.5), loading the aqueous layer to perform ion exchange chromatography at the speed of 1-6.0Bv/h (preferably 4Bv/h), firstly washing by using deionized water, then eluting by using 0.2-4.5M (preferably 1M) of ammonia water at the speed of 0.5-3.0Bv/h (preferably 2Bv/h), detecting the eluent by using filter paper impregnated with 0.2% ninhydrin solution, changing the filter paper to be purple black to indicate that the eluent contains L-amino acid, collecting the eluent containing the target component, distilling under reduced pressure to be pasty, dissolving and recrystallizing by using methanol, taking crystals and drying to obtain the L-amino acid.
The N-phenylacetyl-DL-amino acid is prepared by the following method: adding amino acid and NaOH into distilled water, fully stirring under an ice bath condition of 4 ℃ until the solution is colorless and transparent, dropwise adding phenylacetyl chloride, continuing to react for 2 hours under the ice bath condition of 4 ℃ after dropwise adding is finished, stirring at normal temperature (25-30 ℃) for 5 hours until the solution is colorless and transparent, adding HCl to adjust the pH value to 1.5-5.5 (preferably 2-4), separating out white solid, and performing suction filtration and drying to obtain the N-phenylacetyl-DL-amino acid; the molar ratio of the amino acid to NaOH is 1: 1-7 (preferably 1: 1.5-2.5); the molar ratio of the amino acid to the phenylacetyl chloride is 1: 0.1-2 (preferably 1:0.5-1.0), and the volume dosage of the distilled water is 3-10ml/g based on the mass of the amino acid; the amino acid is one of the following: alanine, serine, glutamic acid, tyrosine, threonine, valine, aspartic acid, methionine, leucine, isoleucine, phenylalanine.
The wet thallus obtained by fermenting the B-resistant lysine bacillus ZJB-17007 is prepared by the following method:
(1) slant culture:
b-lysine-resistant bacillus ZJB-17007 is inoculated to a slant culture medium and cultured for 48h at 30 ℃ to obtain slant thalli; the final concentration of the slant culture medium is as follows: casein peptone 1-20g/L, Na2HPO40.5-5.0g/L,K2HPO40.5-6.0g/L, soybean peptone 1-5g/L, glucose 1-5g/L, NaCL1-10 g/L, agar 20g/L, deionized water as solvent, and pH 6.5-7.5. Preferably, the final concentration composition of the slant culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, 20g/L of agar, deionized water as a solvent and 7.0 of pH value.
(2) Seed culture:
selecting one strain of the thallus on the inclined plane, inoculating the strain to a seed culture medium, and culturing at 30 ℃ for 24 hours to obtain a seed solution; the final concentration of the seed culture medium is as follows: casein peptone 1-20g/L, Na2HPO40.5-5.0g/L,K2HPO40.5-6.0g/L, 1-5g/L of soybean peptone, 1-5g/L of glucose, 1-10g/L of NaCl, deionized water as solvent and 6.5-7.5 of pH value. Preferably, the final concentration composition of the seed culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value.
(3) Fermentation culture:
inoculating the seed solution into a fermentation culture medium in an inoculation amount of 1-10% (preferably 1%) by volume concentration, carrying out shaking culture at 30 ℃ and 150rpm for 60h, centrifuging at 12000g for 10min, and collecting wet thalli; the final concentration composition of the fermentation medium is as follows: casein peptone 1-20g/L, Na2HPO40.5-5.0g/L,K2HPO40.5-6.0g/L, 1-5g/L of soybean peptone, 1-5g/L of glucose, 1-10g/L of NaCl, deionized water as a solvent, and 6.5-7.5 of pH value; preferably, the final concentration of the fermentation medium consists of: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value.
The buffer solution with the pH value of 8.5 comprises the following components: a citric acid buffer (100mM, pH 4.0-6.0), a phosphoric acid buffer (100mM, pH 6.0-8.0), Tris-HCl (100mM, pH 7.0-9.0), a boric acid buffer (100mM, pH 9.0-10.5), an aqueous ammonia buffer (pH 7.0-10.0 of a substrate solution adjusted with aqueous ammonia), a Roche buffer (100mM, pH 10.5-12.0); preferably the buffer composition is: the pH of the substrate solution was adjusted to 8.5 with ammonia.
The invention has the following beneficial effects: the invention provides a bacterial strain capable of producing amidohydrolase, namely lysine bacillus boron tolerance ZJB-17007, and also provides a method for preparing important effective component 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid of L-glufosinate by using wet thallus obtained by fermentation culture of the lysine bacillus boron tolerance ZJB-17007 as a biocatalyst to catalyze 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid, wherein the conversion rate of 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] butyric acid reaches 49.5%, and the product L-glufosinate reaches e.e% 99.9% The method for preparing the L-amino acid from the DL-amino acid has the advantages that the conversion rate can reach 49%, the product chiral amino acid can reach e.e% of 99-99.9%, the resolution preparation process is green and efficient, the strain is easy to culture, collect and apply, the catalytic reaction condition is mild, and the method has an important application prospect.
(IV) description of the drawings
FIG. 1 is a standard curve of OPA/NAC-glufosinate concentration under a high throughput screening method.
FIG. 2 is a liquid phase results diagram of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid and 2-amino-4- [ hydroxy (methyl) phosphoryl ] -D-butyric acid under pre-column derivatization reversed phase high performance liquid chromatography.
FIG. 3 shows the result of electron microscope observation of the strain ZJB-17007 of the screened B-tolerant lysine bacillus (Lysinibacillus boronitolerans).
(V) specific embodiment:
the invention will be further described with reference to specific examples, but the scope of the invention is not limited thereto: the ultrapure water is UP water, and is water with the resistivity of 18M omega cm (25 ℃). The ammonia water is an aqueous solution containing 25-28% of ammonia. The normal temperature of the invention is 25-30 ℃. The ice bath was 4 ℃ in the examples of the invention. The preparation method of the 2% ninhydrin solution comprises the following steps: dissolving 2g of ninhydrin and 0.08g of stannous chloride in 100mL of ultrapure water, stirring and filtering, and taking the filtrate and storing away from light.
Example 1: screening of B.borandii (Lysinibacillus boronitolorans) ZJB-17007
1. Preliminary screening
The invention takes soil samples from all parts of the country, and takes 80 parts of soil samples. The screening method comprises the following specific steps: weighing 1g of soil sample, placing the soil sample in 10 mL0.85% of physiological saline, shaking, standing, taking supernatant into an enrichment medium, and carrying out shaking culture at 30 ℃ and 150r/min for 2-3 days. Adding 1mL of enrichment solution into 50mL of fresh enrichment medium, repeating the process for 3 times, and then carrying out separation and purification.
Bromothymol blue filter paper is selected as indicating filter paper to detect colonies capable of degrading substrates. The principle is as follows: the colonies containing the enzyme of interest, after degradation of the substrate, produce acidic by-products (phenylacetic acid, acetic acid, formic acid, benzoic acid) which cause a change in the pH locally on the indicator filter. The indicator bromothymol blue has the color change range of 5.8-7.6 (yellow-blue), so that the color of the periphery of the colony utilizing chlorine-containing organic matters is yellow, and the color of the plate around the colony which cannot be utilized is green.
Gradually diluting the enriched solution with sterile 0.85% physiological saline for 10 gradients, and selecting 10-4、10-5、10-6、10-7、10-8、10-9、10-100.1mL of each of the five dilutions was spread on the plate screening medium and incubated at 30 ℃ for 48 h. Spreading sterile filter paper (indicator filter paper) soaked with 10% bromothymol blue on the plate, picking out corresponding colony showing yellow on the indicator filter paper, inoculating into 96-well plate containing growth medium, placing into shaker at 30 deg.C and 180rpm for constant temperature culture, during which OD is detected600And (3) drawing a growth curve, transferring 300 microliters of fresh fermentation medium when the strain grows to a logarithmic phase, inoculating the residual strain liquid after the 96-pore plate is inoculated, sealing and storing in a refrigerator at 4 ℃ for later use. And placing the transferred 96-well plate containing the fermentation medium at 30 ℃ and culturing for 48h by a shaking table at 180rpm to ensure that the plate fully grows and produces enzyme to obtain bacterial suspension.
2. OPA/NAC high throughput screening
Dissolving a substrate 2-N-phenylacetyl-4- [ hydroxyl (methyl) phosphoryl ] -DL-butyric acid in ammonia water, adding 1mL of the bacterial suspension prepared in the step 1, adjusting the pH value of the solution to 8.5 by using the ammonia water to form a 10mL reaction system, enabling the final concentration of the substrate to be 50mM, and carrying out constant temperature reaction on a shaker at 30 ℃ and 180rpm for 24 hours. The obtained conversion solution is correspondingly diluted by 10 times by ultrapure water respectively, is subjected to ice bath at 4 ℃, is detected by an OPA/NAC high-throughput screening method, and strains with high relative fluorescence values are screened out.
OPA/NAC high throughput screening method: absorbing 40 mul of diluted ice-bath reaction liquid to a 96-hole fluorescence detection plate by using a line gun, adding the A liquid after ice-bath, oscillating for 30s in an enzyme-labeling instrument, adding 100 mul of ultrapure water, oscillating for 30s again, and carrying out gamma-fluorescence detection on the solution at the lambda positionex=340nm;λem(455 nm) and the fluorescence intensity value obtained was combined with 2-amino-4- [ hydroxy (methyl) phosphoryl group]Comparing the standard curve of the (E) -L-butyric acid to obtain the 2-amino-4- [ hydroxyl (methyl) phosphoryl group contained in the sample]The concentration of L-butyric acid, from which 2-amino-4- [ hydroxy (methyl) phosphoryl can be calculated]The yield of the (E) -L-butyric acid is obtained, and the amidohydrolase is obtained to carry out biocatalytic resolution on the 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl]2-amino-4- [ hydroxy (methyl) phosphoryl group as important effective component for preparing L-glufosinate-ammonium from-DL-butyric acid]-conversion of L-butyric acid.
The solution A comprises: dissolving N-acetyl-L-cysteine (0.448g) and o-phthalaldehyde (0.185g) in 10mL of absolute ethyl alcohol, adding a boric acid buffer solution (140mM, pH 9.5) under an ice bath condition to a constant volume of 50mL, and storing under an ice bath condition for 3 days. The final concentration of N-acetyl-L-cysteine in the solution A is 0.313mol/L, and the final concentration of o-phthalaldehyde is 0.138 mol/L.
The 2-amino-4- [ hydroxy (methyl) phosphoryl group]-L-butyric acid standard curve preparation method: 5g/L of 2-amino-4- [ hydroxy (methyl) phosphoryl group was prepared with ultrapure water]-standard solution of L-butyric acid, diluted in 3 concentration gradients: 0.01-0.1 g/L (0.01, 0.02, 0.04, 0.06, 0.08, 0.1g/L), 0.1-1.0 g/L (0.1, 0.2, 0.4, 0.6, 0.8, 1.0g/L), 1.0-5.0 g/L (1.0, 2.0, 3.0, 4.0, 5.0g/L), respectively absorbing 40 mul to 96-hole micropore fluorescent standard solution with different concentration gradients by a row gun, respectively adding 40 mul of prepared detection plate stored in ice bathOscillating solution A in microplate reader for 30s, adding 100 μ l of ultrapure water, oscillating for 30s, and measuring at λex=340nm;λemThe fluorescence intensity value was measured at 455nm, and the obtained fluorescence intensity value was plotted on the ordinate and the concentration of the standard on the abscissa to prepare a calibration curve. The standard curves of 3 concentration ranges are prepared, and the good linearity (shown in figure 1) can be known when the sample concentration is 0.01-0.1 g/L, R20.9978, showing that the method has high accuracy and applicability.
3. High performance liquid chromatography rescreening
And (3) inoculating the strains screened in the step (2) to a slant culture medium, culturing for 48h at 30 ℃, and storing in a refrigerator at 4 ℃.
The strain preserved on the slant was inoculated into a seed medium and cultured at 30 ℃ for 24 hours. Inoculating the seed liquid into a fermentation medium at an inoculation amount of 1% by volume concentration, and performing shaking culture at 30 ℃ and 150rpm for 60 h. The cells were centrifuged at 12000g for 5min to collect wet cells. Taking wet thalli, adding a substrate of 2-N-phenylacetyl-4- [ hydroxyl (methyl) phosphoryl ] -DL-butyric acid and ammonia water, adjusting the pH of the solution to 8.5 by using the ammonia water to form a 10mL reaction system, wherein the final concentration of the wet thalli is 50g/L, the final concentration of the substrate is 50mM, and placing the reaction system in a water bath shaker at 30 ℃ for conversion for 24h. 1mL of the transformation solution was collected, centrifuged, and the supernatant was analyzed by HPLC. Finally obtaining a wild strain with higher catalytic activity, and marking the strain as a strain ZJB-17007.
The High Performance Liquid Chromatography (HPLC) re-screening method comprises the following steps: centrifuging the conversion solution to obtain a supernatant, diluting the supernatant by 10 times with ultrapure water, taking 200 mu L of clean 1.5mL of EP tube, adding 200 mu L of derivatization reagent, reacting at 30 ℃ for 5min, adding 600 mu L of ultrapure water, mixing uniformly, filtering by using a syringe filter membrane (0.22 mu m), and detecting by HPLC to obtain the concentrations of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid and 2-amino-4- [ hydroxy (methyl) phosphoryl ] -D-butyric acid in the conversion solution.
The derivatization reagent is: 0.1g of o-phthalaldehyde and 0.12g of 0.12g N-phenylacetyl-L-cysteine were dissolved in 10mL of absolute ethanol, and then the solution was diluted to 50mL with a boric acid buffer (100mM, pH 9.8).
The HPLC conditions are as follows: u3000 HPLC and fluorescence detectionA machine; daian C18A silicon hydroxyl packed column (250mM × 4.6.6 mM), a mobile phase of ammonium acetate (50mM pH 4.7) solution containing 10% pure methanol, a column temperature controlled at 35 deg.C, and a fluorescence excitation wavelength lambdaex350nm, emission wavelength λemThe sample size was 10. mu.l at 450 nm. Under these conditions, 2-amino-4- [ hydroxy (methyl) phosphoryl group]-L-butyric acid and 2-amino-4- [ hydroxy (methyl) phosphoryl]The peak time of the-D-butyric acid is 7.800min and 9.290min (as shown in figure 2).
The enrichment medium comprises the following components: 2-N-Phenylacetyl-4- [ hydroxy (methyl) phosphoryl]1g/L of DL-butyric acid, 5g/L of glucose, 1g/L of sodium chloride, K2HPO4·3H2O 0.8g/L,KH2PO43.3g/L,MgSO4·7H2O0.2 g/L, deionized water as solvent, and pH 7.
Plate screening medium composition: 2-N-Phenylacetyl-4- [ hydroxy (methyl) phosphoryl]1g/L of DL-butyric acid, 5g/L of glucose, 1g/L of sodium chloride, K2HPO4·3H2O 0.8g/L,KH2PO43.3g/L,MgSO4·7H20.2g/L of O, 20g/L of agar, deionized water as a solvent and pH 7.
Growth medium composition: 10g/L of sodium chloride, 10g/L of peptone, 5g/L of yeast powder and deionized water as a solvent. The composition of the slant medium, seed medium and fermentation medium was the same as in example 3.
Example 2: identification of Strain ZJB-17007
1. Morphological identification:
the strain ZJB-17007 screened in the embodiment 1 of the invention forms a round or nearly round, soft texture, smooth and flat surface, neat edge and glossy yellowish colony with the diameter of 2-4mm after being cultured for 24 hours at 37 ℃ on a solid culture medium. And (3) gram staining observation: purple long rod shape with spores. Solid medium composition: 10g/L of sodium chloride, 10g/L of peptone, 5g/L of yeast powder, 20g/L of agar and deionized water as a solvent.
2. Physiological and biochemical identification:
94 phenotypic tests were performed on strain ZJB-17007 using the Biolog (GEN III) automated microorganism identification system, including 71 carbon source utilization assays and 23 chemosensitivity assays: inoculating the strain to a specific plate culture medium, culturing at 33 deg.C for 2 days, washing thallus on the plate with sterile cotton swab, mixing with inoculating liquid (IF-A), making into bacterial suspension, and adjusting to 91% T/IF-A with turbidimeter. The bacterial suspensions were added to each well of the BiologGEN iii microwell assay plate using an 8-well electric applicator, 100 μ L per well. The plate was placed in a 33 ℃ incubator and read on a Biolog reader after 12h, 24h, 36h, 48h incubation, respectively.
The strain ZJB-17007 analyzes the metabolic fingerprint through a Biolog reader, the strain ZJB-17007 can strongly utilize 65 carbon sources, and cannot utilize other 3 carbon sources or has weak utilization capacity; the strain ZJB-17007 is sensitive to 23 chemical substances. The 48h identification results given by the Biolog system are shown in tables 1 and 2.
TABLE 1 ability of the Strain ZJB-17007 to utilize 71 carbon sources on the BiologGEN III plate
Figure BDA0001818493090000081
Figure BDA0001818493090000091
TABLE 2 chemosensitivity of Strain ZJB-17007 to 23 chemicals on BiologGEN III plates
Figure BDA0001818493090000092
Molecular biological identification:
the 16S rDNA gene of the strain is amplified by using the total DNA of the strain ZJB-17007 as a template and using primers P1:5'-AGAGTTTGATCCTGGCTCAG-3' and P2:5'-AAGGAGGTGATCCAGCCGCA-3', after the gene product is connected with a T vector, Shanghai workers are entrusted to amplify and sequence the 16S rDNA of the strain to obtain a 16S rDNA sequence (SEQ ID NO.1) of the strain, the 16S rDNA gene sequences of related strains in GenBank are searched by BLAST on an NCBI website, and homology comparison is carried out. The microorganism of the invention has the highest homology (homology, 99 percent, based on 16S ribosomal RNA gene) with the Lysinibacillus boronitolerans strain NBRC 103108, and the strain is basically identified to belong to a control strain based on the 16S rDNA homology higher than 95 percent according to the principle of microbial genetic identification. Therefore, the strain ZJB-17007 identified by the experiment is boron-resistant lysine bacillus (Lysinibacillus boronitorens), is named as boron-resistant lysine bacillus (Lysinibacillus boronitorens) ZJB-17007, is preserved in the China center for type culture Collection, and has the preservation number of CCTCC No: m2017602, date of deposit 2017, 10 month 23 day, address: university of Wuhan, China 430072.
Any nucleotide sequence obtained by substituting, deleting or inserting one or more nucleotides into the nucleotide sequence shown in SEQ ID NO.1 in the nucleotide sequence table is within the protection scope of the present invention as long as the nucleotide sequence has homology of more than 90%.
Example 3: preparation of Wet cells
(1) Slant culture:
b-lysine-resistant bacillus ZJB-17007 is inoculated to a slant culture medium and cultured for 48h at 30 ℃ to obtain slant thalli;
the final concentration of the slant culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, 20g/L of agar, deionized water as a solvent and 7.0 of pH value.
(2) Seed culture:
selecting one strain of the thallus on the inclined plane, inoculating the strain to a seed culture medium, and culturing at 30 ℃ for 24 hours to obtain a seed solution;
the final concentration of the seed culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value.
(3) Fermentation culture:
inoculating the seed solution into a fermentation culture medium in an inoculation amount of 1% of volume concentration, carrying out shaking culture at 30 ℃ and 150rpm for 60h, centrifuging at 12000g for 10min, and collecting wet thalli;
the hairThe final concentration of the fermentation medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value.
Example 4 bioconversion reactions Using 2-N-Phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
(1) The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 0.3g of the wet cell prepared in example 3 was added thereto, and the pH was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, and the resulting solution was placed in a 30 ℃ water bath shaker at 150rpm for 24 hours to convert the substrate to a final concentration of 50 mM. 1mL of the transformation solution was added to an ep tube, centrifuged, and the supernatant was analyzed and assayed by HPLC as described in example 1. The results show that ZJB-17007 can convert the substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid into the product 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid, the conversion rate is 49.7%, and the optical purity of the product 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid is 99.9%.
(2) Separating the conversion liquid containing the product 2-amino-4- [ hydroxyl (methyl) phosphoryl ] -L-butyric acid obtained by the enzyme catalytic reaction to extract the product 2-amino-4- [ hydroxyl (methyl) phosphoryl ] -L-butyric acid.
1) And (3) extraction:
the resulting conversion solution was added to a separatory funnel, and simultaneously, dichloromethane of equal volume was added, and after shaking, it was allowed to stand for 3 hours, the organic layer was discharged from the lower end of the separatory funnel, and the aqueous layer was poured out from the upper end. The aqueous layer obtained is an aqueous solution containing the product 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid.
2) Product separation using anion exchange resins
(ii) resin pretreatment
Soaking resin 201 × 7 in 50 deg.C warm water to swell the resin and remove fine particles (inclined or flotation method), soaking in 1.0M NaOH water solution for 3h, washing with deionized water to neutral, soaking in 1.0M HCl water solution for 3h, washing with deionized water to neutral, soaking in 1.0M NaOH water solution for 3h, and converting into OH-Form, finally useWashing with ion water to neutrality for use.
② mounting columns
The method comprises the steps of filling a column (with the inner diameter of 1.5cm and the height of 40cm) by a wet method, firstly adding deionized water with the height of 13cm into an ion exchange column, then filling 30mL of wet resin 201 multiplied by 7 into a glass cup, adding 50mL of deionized water, slowly stirring, pouring suspended resin into the ion exchange column, naturally settling, and enabling the resin to be uniformly secreted in the column without obvious boundary lines, bubbles and the like.
③ sample loading and elution
Adjusting the pH of the aqueous solution obtained in the step 1) to 2.5, loading the aqueous solution at the column flow rate of 4.0Bv/h, taking out effluent liquid at intervals for liquid phase detection, when the adsorption reaches the maximum value, washing the effluent liquid with deionized water, then eluting the effluent liquid with 1.0M ammonia water at the elution speed of 2.0Bv/h, detecting the effluent liquid with filter paper impregnated with 0.2% ninhydrin solution, wherein the filter paper changes purple to show that the eluent contains the target substance 2-amino-4- [ hydroxy (methyl) phosphoryl group]L-butyric acid, collecting the eluate containing the target component, and detecting the 2-amino-4- [ hydroxy (methyl) phosphoryl group contained therein at intervals]-the content of L-butyric acid. Washing the ion exchange column with deionized water after the elution is finished, and converting the resin into OH-For the next separation.
Purification
Distilling the eluent under reduced pressure to obtain a yellow viscous substance, adding methanol to dissolve the yellow viscous substance, stirring the yellow viscous substance under ice bath to recrystallize the yellow viscous substance to obtain a white solid, and filtering the white solid to obtain the product 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid.
After 1L of the conversion solution prepared under the reaction condition of the step (1) is separated and purified by the method, 4.9g of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid solid is obtained, the yield of the product is 98.9%, and the optical purity is 99.9%.
Example 5: biotransformation reaction using 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 0.5g of the wet cell prepared in example 3 was added thereto, and the pH was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 100mM, and the reaction system was placed in a 30 ℃ water bath shaker and inverted at 150rpm for 24 hours. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.9%. 1L of the converted solution was prepared under the same reaction conditions and separated and purified by the method in example 4 to obtain 9.2g of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid, the yield of which was 92.9% and the optical purity was 99.9%.
Example 6: biotransformation reaction using 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 0.6g of the wet cell prepared in example 3 was added thereto, and the pH was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 200mM, and the reaction system was placed in a 30 ℃ water bath shaker and inverted at 150rpm for 24 hours. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.9%. 1L of the converted solution was prepared under the same reaction conditions and separated and purified by the method in example 4 to obtain 19.0g of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid, the yield of which was 95.9% and the optical purity of which was 99.9%.
Example 7: biotransformation reaction using 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 2g of the wet cell prepared in example 3 was added thereto, and the pH was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 200mM, and the reaction system was placed in a 30 ℃ water bath shaker and inverted at 150rpm for 24 hours. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.9%. 1L of the converted solution was prepared under the same reaction conditions and separated and purified by the method in example 4 to obtain 19.5g of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid, the yield of which was 97.9% and the optical purity was 99.9%.
Example 8: biotransformation reaction using 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 0.5g of the wet cell prepared in example 3 was added thereto, and the pH was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 50mM, the reaction system was placed in a 40 ℃ water bath shaker, and the resultant was transformed at 150rpm for 24 hours, and 1mL of the transformed solution was taken out into an EP tube, and after centrifugation, the supernatant was subjected to HPLC analysis as described in example 1 to determine the conversion rate to be 49.9%. 1L of the converted solution was prepared under the same reaction conditions, and after separation and purification by the method in example 4, 4.86g of the product 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid was obtained, with a yield of 98.1% and an optical purity of 99.9%.
Example 9: biotransformation reaction using 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid as substrate
The substrate 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid was dissolved in aqueous ammonia, 2g of the wet cell prepared in example 3 was added thereto, pH8.5 was adjusted with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 200mM, the reaction system was placed in a 50 ℃ water bath shaker, and the shaking was carried out at 150rpm for 24 hours, 1mL of the conversion solution was taken out into an EP tube, and after centrifugation, the supernatant was subjected to HPLC analysis as described in example 1 to obtain a conversion rate of 49.9%. 1L of the converted solution was prepared under the same reaction conditions and separated and purified by the method in example 4 to obtain 19.4g of 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid, the yield of which was 97.9% and the optical purity was 99.9%.
Example 10: biotransformation reaction using N-phenylacetyl-DL-alanine as substrate
Adding 6.0g of alanine and 4.6g of NaOH into 30ml of distilled water, fully stirring under an ice bath condition until the solution is colorless and transparent, dropwise adding 5.0g of phenylacetyl chloride, after the dropwise adding is completed, the solution is light yellow, continuing to react for 2 hours under the ice bath condition, stirring at normal temperature for reaction for 5 hours until the solution is colorless and transparent, adding HCl to adjust the pH value to about 2.0, separating out a white solid, and performing suction filtration and drying to obtain 13g of white solid namely the N-phenylacetyl-DL-alanine.
Substrate N-phenylacetyl-DL-alanine was dissolved in aqueous ammonia, 0.6g of wet cell prepared in example 3 was added thereto, pH8.5 was adjusted with aqueous ammonia to prepare 10mL of a reaction system, and the reaction system was stirred in a 30 ℃ water bath shaker at 150rpm for 24 hours with the substrate added to a final concentration of 100 mM. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.9%. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was performed with filter paper impregnated with 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-alanine was obtained in an amount of 4.36g, the yield was 97.9%, and the optical purity was 99.9%.
Example 11: biotransformation reaction using N-phenylacetyl-DL-threonine as substrate
Adding 6.0g of threonine and 4.6g of NaOH into 30ml of distilled water, fully stirring under an ice bath condition until the solution is colorless and transparent, dropwise adding 5.0g of phenylacetyl chloride, after dropwise adding is completed, keeping the solution in light yellow, reacting for 2 hours under the ice bath condition, stirring at normal temperature for reaction for 5 hours until the solution is colorless and transparent, adding HCl to adjust the pH value to about 2.0, separating out a white solid, and performing suction filtration and drying to obtain the white solid, namely 10.35g of N-phenylacetyl-DL-threonine.
Substrate N-phenylacetyl-DL-threonine was dissolved in aqueous ammonia, 3g of the wet cell prepared in example 3 was added thereto, pH8.5 was adjusted with aqueous ammonia to prepare 100mL of a reaction system, and the reaction system was stirred in a 30 ℃ water bath shaker at 150rpm for 24 hours with the substrate added to a final concentration of 100 mM. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.9%. 1L of the conversion solution was prepared under the same reaction conditions, and after separation and purification by the method in example 4 (detection was carried out with filter paper impregnated with 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-threonine was obtained in an amount of 5.88g, with a yield of 98.8% and an optical purity of 99.9%.
Example 12: biotransformation reaction using N-phenylacetyl-DL-tyrosine as substrate
3.619g of DL-tyrosine and 1.8g of NaOH are added into 25ml of distilled water, the mixture is fully stirred under the ice bath condition until the solution is transparent, 2.875g of phenylacetyl chloride is dropwise added, the solution is milky after the dropwise addition, the ice bath condition is continued for reaction for 2h, the mixture is stirred at normal temperature for reaction for 5h, HCl is added for regulating the pH value to about 2.0, a large amount of white solid is separated out, and the white solid is 5.5g of N-phenylacetyl-DL-tyrosine after suction filtration and drying.
Substrate N-phenylacetyl-DL-tyrosine was dissolved in ammonia water, 0.3g of wet cell prepared in example 3 was added thereto, pH8.5 was adjusted with ammonia water to prepare a 10mL reaction system, and the reaction system was stirred in a 30 ℃ water bath shaker at 150rpm for 24 hours with the substrate added to a final concentration of 100 mM. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain 49.8% conversion. After 1L of the transformant was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was carried out using a filter paper impregnated with a 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-tyrosine 8.84g was obtained, the yield of the product was 97.6%, and the optical purity was 99.9%.
Example 13: biotransformation reaction using N-phenylacetyl DL-phenylalanine as substrate
Adding 3.3g of DL-phenylalanine and 1.8g of NaOH into 25ml of distilled water, fully stirring under an ice bath condition, thoroughly brightening the solution, dropwise adding 2.875g of phenylacetyl chloride, reacting for 2 hours under the ice bath condition after dropwise adding is finished, adding HCl to adjust the pH value to about 4.0 after stirring and reacting for 5 hours at normal temperature, separating out a large amount of white solid, and performing suction filtration and drying to obtain 5.2g of white solid, namely N-phenylacetyl-DL-phenylalanine.
Substrate N-phenylacetyl-DL-phenylalanine was dissolved in ammonia water, 0.4g of wet cell prepared in example 3 was added thereto, pH8.5 was adjusted with ammonia water to prepare 10mL of a reaction system, and the reaction system was stirred in a 30 ℃ water bath shaker at 150rpm for 24 hours with the substrate added to a final concentration of 100 mM. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain 49.8% conversion. 1L of the reaction solution was prepared under the same reaction conditions, and after separation and purification by the method in example 4 (detection was performed with filter paper impregnated with 0.2% ninhydrin solution, and the filter paper turned purple-black indicating that the eluate contained the target substance), the product L-phenylalanine 8.11g was obtained, the product yield was 98.3%, and the optical purity was 99.9%.
Example 14: biotransformation reaction using N-phenylacetyl-DL-leucine as substrate
Adding 3.92g of leucine and 3.2g of NaOH into 30ml of distilled water, fully stirring under an ice bath condition until the solution is bright, dropwise adding 6.2g of phenylacetyl chloride, reacting for 2 hours under the ice bath condition continuously, stirring at normal temperature for 5 hours, adding HCl to adjust the pH value to about 2.0, precipitating a large amount of white solid, and performing suction filtration and drying to obtain 7.43g of white solid, namely N-phenylacetyl-DL-leucine.
Substrate N-phenylacetyl-DL-leucine was dissolved in ammonia water, 0.6g of wet cells prepared in example 3 was added thereto, pH8.5 was adjusted with ammonia water to prepare 10mL of a reaction system, the substrate was added to a final concentration of 100mM, the mixture was placed in a 30 ℃ water bath shaker, and transformed at 150rpm for 24 hours, 1mL of the transformed solution was taken out into an EP tube, and after centrifugation, the supernatant was collected and subjected to HPLC analysis as described in example 1 to determine the conversion rate to be 49.7%. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was performed with filter paper impregnated with 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-leucine (6.32 g) was obtained, the yield of the product was 96.4%, and the optical purity was 99.9%.
Example 15: biotransformation reaction using N-phenylacetyl-DL-isoleucine as substrate
Adding 7.84g of isoleucine and 6.4g of NaOH into 60ml of distilled water, fully stirring under an ice bath condition, dripping 5.78g of phenylacetyl chloride into the solution to obtain a light yellow solution, continuing to react for 2 hours under the ice bath condition, stirring at normal temperature for 5 hours, adding HCl to adjust the pH value to about 2.0, enabling the solution to become colorless and transparent, separating out a small amount of white solid, and performing suction filtration and drying to obtain 7.28g of white solid, namely N-phenylacetyl-DL-isoleucine.
Dissolving a substrate N-phenylacetyl-DL-isoleucine in ammonia water, adding 0.5g of the wet bacterial cells prepared in the method in example 3, adjusting pH to 8.5 with ammonia water to form a 10mL reaction system, adding the substrate to a final concentration of 100mM, placing the mixture in a water bath shaker at 30 ℃, converting the mixture at 150rpm for 24h, taking 1mL of the conversion solution to an EP tube, centrifuging the mixture, and taking the supernatant to perform HPLC analysis according to the method in example 1 to detect that the conversion rate is 49.8%. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 1 (detection was carried out using filter paper impregnated with 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-isoleucine was obtained in an amount of 6.41g, the yield was 93.7%, and the optical purity was 99.9%.
Example 16: biotransformation reaction using N-phenylacetyl-DL-serine as substrate
Adding 6.0g of serine and 4.6g of NaOH into 20ml of distilled water, fully stirring under an ice bath condition until the solution is colorless and transparent, dropwise adding 5.2g of phenylacetyl chloride, after dropwise adding is completed, keeping the solution in light yellow, continuing to react for 2 hours under the ice bath condition, stirring at normal temperature for 5 hours until the solution is colorless and transparent, adding HCl to adjust the pH value to about 2.0, separating out a white solid, and performing suction filtration and drying to obtain 11g of white solid, namely N-phenylacetyl-DL-serine.
Dissolving a substrate N-phenylacetyl-DL-serine in ammonia water, adding 0.5g of wet thalli prepared by the method in example 3, adjusting the pH of the solution to 8.5 by using the ammonia water to form a 10mL reaction system, adding the substrate to the solution with the final concentration of 100mM, placing the solution in a water bath shaker at 30 ℃, converting the solution at 150rpm for 24 hours, taking 1mL of conversion solution to an EP tube, centrifuging the solution, taking the supernatant, and performing HPLC analysis according to the method in example 1 to detect the conversion rate of 49.5 percent. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was carried out using filter paper impregnated with 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-serine (4.91 g) was obtained, the yield was 93.5%, and the optical purity was 99.9%.
Example 17: biotransformation reaction using N-phenylacetyl-DL-methionine as substrate
Adding 8.94g of methionine and 7.2g of NaOH into 120ml of distilled water, fully stirring under an ice bath condition until the solution is transparent, dropwise adding 10.3g of phenylacetyl chloride, after dropwise adding is completed, the solution is transparent, continuing to react for 2 hours under the ice bath condition, stirring at normal temperature for 5 hours, adding HCl to adjust the pH value to about 2.0, separating out a large amount of white solid, and performing reduced pressure suction filtration and drying to obtain 14.8g of N-phenylacetyl-DL-methionine.
Substrate N-phenylacetyl-DL-methionine was dissolved in ammonia, 0.45g of wet cell prepared in example 3 was added thereto, and the pH of the solution was adjusted to 8.5 with ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 100mM, and the reaction system was placed in a 30 ℃ water bath shaker and inverted at 150rpm for 24 hours. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.4%. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was carried out using a filter paper impregnated with a 0.2% ninhydrin solution, and the purplish black color of the filter paper indicated that the eluate contained the target substance), the product L-methionine (7.22 g) was obtained, the yield of the product was 96.9%, and the optical purity was 99.9%.
Example 18: biotransformation reaction using N-phenylacetyl-DL-valine as substrate
9.36g of valine and 7.2g of NaOH are added into 60ml of distilled water, the mixture is fully stirred under the ice bath condition until the solution is transparent, 14.24g of phenylacetyl chloride is dropwise added, the solution is yellowish and slightly turbid after the dropwise addition, the mixture is continuously reacted for 2 hours under the ice bath condition, the solution is colorless and transparent after being stirred at normal temperature for 5 hours, HCl is added to adjust the pH value to about 2.0, a large amount of white solid is separated out, and the white solid, namely 17.3g of N-phenylacetyl-DL-valine, is obtained after suction filtration and drying.
Substrate N-phenylacetyl-DL-valine was dissolved in aqueous ammonia, 0.2g of the wet cell prepared in example 3 was added thereto, and the pH of the solution was adjusted to 8.5 with aqueous ammonia to prepare 10mL of a reaction system, wherein the substrate was added to a final concentration of 100mM, and the reaction system was placed in a 30 ℃ water bath shaker and inverted at 150rpm for 24 hours. 1mL of the transformation solution was transferred to an EP tube, centrifuged, and the supernatant was analyzed by HPLC as described in example 1 to obtain a conversion of 49.2%. 1L of the converted solution was prepared under the same reaction conditions, and after separation and purification by the method in example 4 (detection was carried out using a filter paper impregnated with a 0.2% ninhydrin solution, and the filter paper became purple-black indicating that the eluate contained the target substance), the product L-valine (5.62 g) was obtained, the yield of the product was 96.0%, and the optical purity was 99.9%.
Example 19: biotransformation reaction using N-phenylacetyl-DL-glutamic acid as substrate
Adding 5g of glutamic acid and 4.6g of NaOH into 30ml of distilled water, fully stirring under an ice bath condition until the solution is transparent, dripping 5.735g of phenylacetyl chloride, after the dripping is finished, the solution is transparent, continuing to react for 2 hours under the ice bath condition, stirring at normal temperature for reaction for 5 hours, adding HCl to adjust the pH value to about 2.0, enabling the solution to become turbid, refrigerating overnight at 4 ℃ in a refrigerator, precipitating a large amount of white solid, and performing reduced pressure suction filtration and drying to obtain 8.1g of N-phenylacetyl-DL-glutamic acid.
Dissolving a substrate N-phenylacetyl-DL-glutamic acid in ammonia water, adding 0.4g of wet thallus prepared by the method in example 3, adjusting the pH of the solution to 8.5 by using the ammonia water to form a 10mL reaction system, adding the substrate to the solution with the final concentration of 100mM, placing the solution in a water bath shaker at 30 ℃, converting the solution at 150rpm for 24 hours, taking 1mL of conversion solution to an ep tube, centrifuging, taking the supernatant, and performing HPLC analysis according to the method in example 1 to detect the conversion rate of 49.3%. After 1L of the conversion solution was prepared under the same reaction conditions and separated and purified by the method in example 4 (detection was performed with filter paper impregnated with 0.2% ninhydrin solution, and the filter paper turned purple-black indicating that the eluate contained the target substance), the product L-glutamic acid (7.05 g) was obtained, the yield of the product was 95.9%, and the optical purity was 99.9%.
Sequence listing
<110> Zhejiang industrial university
<120> B-resistant lysine bacillus ZJB-17007 and application thereof
<160>1
<170>SIPOSequenceListing 1.0
<210>1
<211>1408
<212>DNA
<213> B-tolerant lysine bacillus (Lysinibacillus boronolans)
<400>1
ccttcggcgg ctggctccaa aggttacctc accgacttcg ggtgttacaa actctcgtgg 60
tgtgacgggc ggtgtgtaca aggcccggga acgtattcac cgcggcatgc tgatccgcga 120
ttactagcga ttccggcttc atgtaggcga gttgcagcct acaatccgaa ctgagaacga 180
ctttatcgga ttagctccct ctcgcgagtt ggcaaccgtt tgtatcgtcc attgtagcac 240
gtgtgtagcc caggtcataa ggggcatgat gatttgacgt catccccacc ttcctccggt 300
ttgtcaccgg cagtcacctt agagtgccca actaaatgat ggcaactaag atcaagggtt 360
gcgctcgttg cgggacttaa cccaacatct cacgacacga gctgacgaca accatgcacc 420
acctgtcacc gttgcccccg aaggggaaac tatatctcta cagtggtcaa cgggatgtca 480
agacctggta aggttcttcg cgttgcttcg aattaaacca catgctccac cgcttgtgcg 540
ggcccccgtc aattcctttg agtttcagtc ttgcgaccgt actccccagg cggagtgctt 600
aatgcgttag ctgcagcact aaggggcgga aaccccctaa cacttagcac tcatcgttta 660
cggcgtggac taccagggta tctaatcctg tttgctcccc acgctttcgc gcctcagcgt 720
cagttacaga ccagaaagtc gccttcgcca ctggtgttcc tccaaatctc tacgcatttc 780
accgctacac ttggaattcc actttcctct tctgcactca agtcccccag tttccaatga 840
ccctccacgg ttgagccgtg ggctttcaca tcagacttaa aggaccgcct gcgcgcgctt 900
tacgcccaat aattccggac aacgcttgcc acctacgtat taccgcggct gctggcacgt 960
agttagccgt ggctttctaa taaggtaccg tcaaggtaca gccagttact actgtacttg 1020
ttcttccctt acaacagagt tttacgatcc gaaaaccttc ttcactcacg cggcgttgct 1080
ccatcaggct ttcgcccatt gtggaagatt ccctactgct gcctcccgta ggagtctggg 1140
ccgtgtctca gtcccagtgt ggccgatcac cctctcaggt cggctacgca tcgtcgcctt 1200
ggtgagccgt tacctcacca actagctaat gcgccgcggg cccatcctat agcgacagcc 1260
gaaaccgtct ttcagtctct cgccatgaag caagagagat tattcggtat tagccccggt 1320
ttcccggagt tatcccaaac tatagggtag gttgcccacg tgttactcac ccgtccgccg 1380
ctaacgtcaa aggagcaagc tccttttc 1408

Claims (10)

1. Boron-resistant lysine bacillus (lysine bacillus boronitolerans) ZJB-17007, which is preserved in China center for type culture Collection with the preservation number of CCTCC No: m2017602, date of deposit 2017, 10 month 23 day, address: china, Wuhan university, zip code 430072.
2. The use of the B-tolerant lysinibacillus kZJB-17007 according to claim 1 in catalyzing 2-N-phenylacetyl-4- [ hydroxy (methyl) phosphoryl ] -DL-butyric acid to prepare 2-amino-4- [ hydroxy (methyl) phosphoryl ] -L-butyric acid.
3. Use according to claim 2, characterized in that: the application takes wet thallus obtained by fermenting and culturing the B-lysine-resistant bacillus ZJB-17007 as a catalyst, 2-N-phenylacetyl-4- [ hydroxyl (methyl) phosphoryl ] -DL-butyric acid as a substrate and a buffer solution as a reaction medium to form a reaction system with the pH of 8.5, the conversion reaction is carried out at the temperature of 25-55 ℃ and the speed of 100-200rpm, and after the reaction is finished, the reaction solution is separated and purified to obtain the 2-amino-4- [ hydroxyl (methyl) phosphoryl ] -L-butyric acid.
4. Use according to claim 3, characterized in that: in the reaction system, the dosage of the catalyst is 10-200g/L calculated by the weight of wet thalli, and the initial adding final concentration of the substrate is 10-500 mM.
5. Use according to claim 3, characterized in that: the method for separating and purifying the reaction liquid comprises the following steps: after the reaction is finished, extracting the reaction liquid by using dichloromethane, adjusting the pH value of a water layer to 1.0-5.0, loading the water layer to perform ion exchange chromatography at the speed of 1-6.0Bv/h, washing the water layer by using deionized water, eluting the water layer by using 0.2-4.5M ammonia water at the speed of 0.5-3.0Bv/h, collecting eluent containing the target component, distilling the eluent under reduced pressure to paste, dissolving the paste by using methanol for recrystallization, taking crystals and drying the crystals to obtain the 2-amino-4- [ hydroxyl (methyl) phosphoryl ] -L-butyric acid.
6. The use of the boron-tolerant lysine bacillus ZJB-17007 of claim 1 to catalyze N-phenylacetyl-DL-amino acids to produce N-phenylacetyl-L-amino acids.
7. The use of claim 6, wherein: the application takes wet thallus obtained by fermentation culture of the boron-resistant lysine bacillus ZJB-17007 as a catalyst, takes N-phenylacetyl-DL-amino acid as a substrate, takes a buffer solution as a reaction medium to form a conversion system with the pH of 8.5, carries out conversion reaction at the temperature of 25-55 ℃ and the speed of 100-200rpm, and after the reaction is finished, the reaction solution is separated and purified to obtain the N-phenylacetyl-L-amino acid.
8. The use of claim 7, wherein: in the conversion system, the dosage of the catalyst is 20-300g/L calculated by the weight of wet bacteria, and the initial adding final concentration of the substrate is 50-500 mM.
9. The use of claim 7, wherein: the N-phenylacetyl-DL-amino acid is one of the following: N-phenylacetyl-DL-alanine, N-phenylacetyl-DL-serine, N-phenylacetyl-DL-glutamic acid, N-phenylacetyl-DL-tyrosine, N-phenylacetyl-DL-threonine, N-phenylacetyl-DL-valine, N-phenylacetyl-DL-aspartic acid, N-phenylacetyl-DL-methionine, N-phenylacetyl-DL-leucine, N-phenylacetyl-DL-isoleucine, N-phenylacetyl-DL-phenylalanine.
10. The use of claim 7, wherein: the wet thallus is prepared by the following method: (1) slant culture: b-lysine-resistant bacillus ZJB-17007 is inoculated to a slant culture medium and cultured for 48h at 30 ℃ to obtain slant thalli; the final concentration of the slant culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, 20g/L of agar, deionized water as a solvent and 7.0 of pH value;
(2) seed culture: selecting one strain of the thallus on the inclined plane, inoculating the strain to a seed culture medium, and culturing at 30 ℃ for 24 hours to obtain a seed solution; the final concentration of the seed culture medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value;
(3) fermentation culture:inoculating the seed solution into a fermentation culture medium in an inoculation amount with the volume concentration of 1-10%, carrying out shaking culture at 30 ℃ and 150rpm for 60h, centrifuging at 12000g for 10min, and collecting wet thalli; the final concentration composition of the fermentation medium is as follows: casein peptone 17g/L, Na2HPO43.0g/L,K2HPO41.5g/L, 3g/L of soybean peptone, 2.5g/L of glucose, 5g/L of NaCl, deionized water as a solvent and 7.0 of pH value.
CN201811153717.4A 2018-09-30 2018-09-30 B-lysine-resistant bacillus ZJB-17007 and application thereof Active CN109321493B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811153717.4A CN109321493B (en) 2018-09-30 2018-09-30 B-lysine-resistant bacillus ZJB-17007 and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811153717.4A CN109321493B (en) 2018-09-30 2018-09-30 B-lysine-resistant bacillus ZJB-17007 and application thereof

Publications (2)

Publication Number Publication Date
CN109321493A CN109321493A (en) 2019-02-12
CN109321493B true CN109321493B (en) 2020-10-02

Family

ID=65266400

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811153717.4A Active CN109321493B (en) 2018-09-30 2018-09-30 B-lysine-resistant bacillus ZJB-17007 and application thereof

Country Status (1)

Country Link
CN (1) CN109321493B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111187726B (en) * 2019-10-11 2021-06-22 海南大学 Rice blast bactericide prepared by using lysine-resistant bacillus borreliensis as chassis cells
CN111100806B (en) * 2019-10-11 2022-12-02 海南大学 Areca-nut root rot bactericide prepared by taking boron-resistant lysine bacillus as underpan cells
CN113817628B (en) * 2021-07-20 2023-03-14 海南大学 Mango stem rot bactericide and application thereof
CN114209811B (en) * 2021-12-15 2024-03-29 海南大学 Preparation method of boron-resistant lysine bacillus bacteriocin analogue and application of boron-resistant lysine bacillus bacteriocin analogue in inhibition of bacillus pumilus

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2888449C (en) * 2012-11-15 2021-01-19 Peter INNES Mulch and potting soil compositions containing microorganisms and related methods
CN103275896B (en) * 2013-05-27 2015-03-04 浙江工业大学 Bacillus cereus and application for Bacillus cereus in preparation for L-glufosinate via microbial transformation
CN107118976B (en) * 2017-03-31 2020-04-21 浙江工业大学 Enterobacter cloacae and application thereof
CN106978368B (en) * 2017-03-31 2020-04-21 浙江工业大学 Raoultella ornithinolytica and application thereof

Also Published As

Publication number Publication date
CN109321493A (en) 2019-02-12

Similar Documents

Publication Publication Date Title
CN109321493B (en) B-lysine-resistant bacillus ZJB-17007 and application thereof
CN109609475A (en) Glufosinate-ammonium dehydrogenase mutant and its application for synthesizing L-glufosinate-ammonium
CN107118976B (en) Enterobacter cloacae and application thereof
CN109762768B (en) Bacillus B8W22 and application thereof
CN109321494B (en) Kluyveromyces intermedia ZJB-17004 and application thereof
CN101033454A (en) Erythro micrococcus ZJB-064 and application thereof
CN109182200B (en) Lysinibacillus fusiformis ZJB-17006 and application thereof
CN109321490B (en) Enterobacter aerogenes ZJB-17003 and application thereof
CN109182201B (en) Enterobacter hesheng biological I type ZJB-17002 and application thereof
CN109321492B (en) Non-decarboxylation lechler bacterium ZJB-17008 and application thereof
CN109321491B (en) Xiangfang enterobacter ZJB-17001 and application thereof
CN109321497B (en) Lysine-elongated bacillus ZJB-17009 and application thereof
CN101952418A (en) Produce (2S, 3R, 4S)-method of 4-hydroxy-L-isoleucine
CN109321495B (en) Clinkerwell ZJB-17005 and application thereof
CN109321496B (en) Citrobacter freundii ZJB-17010 and application thereof
CN107142227B (en) Acid-producing klebsiella and application thereof
CN115786315A (en) Acylase, coding gene, engineering bacterium and application in hydrolyzing and synthesizing N-fatty acyl-glutamic acid type surfactant
KR100352192B1 (en) A New thermophilic bacterium Brevibacillus borstelensis BCS-1 and A thermostable D-stereospecific amino acid amidase produced therefrom
CN111286470A (en) Bacillus subtilis capable of degrading erythromycin and application thereof
Kato et al. Enzymatic synthesis of l-β-chloroalanine using amino acid dehydrogenase
CN110804602A (en) L-aspartic acid β -decarboxylase mutant and application thereof
Gul et al. Fermentative production of glutamate by newly isolated soil bacteria
CN109097374A (en) A kind of preparation method, bacterial strain and its application of pseudomonas aeruginosa engineered strain
CN116064302B (en) Vibrio bottom desulfurization SG127 and application thereof
CN116004470B (en) Helicobacter sulfide SG202 and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant