CN113881728B - Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1) - Google Patents

Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1) Download PDF

Info

Publication number
CN113881728B
CN113881728B CN202111166689.1A CN202111166689A CN113881728B CN 113881728 B CN113881728 B CN 113881728B CN 202111166689 A CN202111166689 A CN 202111166689A CN 113881728 B CN113881728 B CN 113881728B
Authority
CN
China
Prior art keywords
leu
ala
deazaguanine
arg
asp
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
CN202111166689.1A
Other languages
Chinese (zh)
Other versions
CN113881728A (en
Inventor
赵弘
于铁妹
潘俊锋
刘建
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.)
Shenzhen Readline Biotechnology Co ltd
Original Assignee
Shenzhen Readline Biotechnology Co ltd
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 Shenzhen Readline Biotechnology Co ltd filed Critical Shenzhen Readline Biotechnology Co ltd
Priority to CN202111166689.1A priority Critical patent/CN113881728B/en
Publication of CN113881728A publication Critical patent/CN113881728A/en
Application granted granted Critical
Publication of CN113881728B publication Critical patent/CN113881728B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/16Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms containing two or more hetero rings
    • C12P17/165Heterorings having nitrogen atoms as the only ring heteroatoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The invention relates to the field of nucleoside preparation, in particular to a preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1). Aiming at the advantages and defects of the preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1) by enzyme catalysis and chemical synthesis, the invention develops a method for preparing the chemical enzyme combination, which shortens the preparation route, greatly improves the conversion efficiency of each step, and greatly reduces the waste discharge of the whole preparation route due to the participation of enzyme catalysis, thereby improving the safety and green index in the industrial production while ensuring the market competitiveness.

Description

Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)
Technical Field
The invention relates to the field of nucleoside preparation, in particular to a preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1).
Background
7-aminomethyl-7-deazaguanine (PreQ 1) is an important starting material for the synthesis of Q nucleosides (queuosine), a highly modified nucleoside similar to guanidine, which are widely found in certain tRNA's of bacteria and eukaryotes and are reported to be preferentially used in t-RNA synthesis, but the intrinsic mechanism of which is not fully understood. Q nucleosides are reported to have a coenzyme effect in prokaryotes and can act potentially as probiotics (growth promoting substances) in higher animals. Q nucleoside is a novel compound produced by microorganism-host interaction, and cannot be synthesized by human body, and its source is mainly human symbiotic microorganism. The base of the Q nucleoside is called Q base (queuine), the main constituent of which is 7-aminomethyl-7-deazaguanine (PreQ 1). 7-aminomethyl-7-deazaguanine is an essential starting material, both in the biosynthesis and chemical preparation of Q nucleosides.
Existing methods for preparing PreQ1 include enzymatic and chemical synthesis methods:
enzymatic method: in the in vivo synthesis of preQ1, five steps of enzyme (GCHI/QueD/QueE/QueF) transformation are required to obtain preQ1 using Guanosine Triphosphate (GTP) as a starting material. Therefore, the route is long, the overall yield is low, and no scale-up preparation using this method has been reported so far.
Chemical synthesis method: there is also little research on the chemical preparation of PreQ1, but a large amount of functional group protection, deprotection, and complicated chemical transformations are required to achieve (more than 7 steps of chemical reactions are required), and the industrial preparation possibility is low.
The preparation route of the 7-aminomethyl-7-deazapurine enzyme method is too long, and an effective in-vitro enzyme catalysis process is difficult to realize; the chemical preparation of the catalyst needs to protect and deprotect a plurality of functional groups, the whole route is long, and various toxic and harmful reagents (such as pivaloyl chloride, TMSTF, cesium acetate and the like) are needed in the chemical preparation process, so that the safety coefficient in the production process is low, the environmental compatibility is low, and the whole yield is not high; the final production cost is high.
With public emphasis on personal safety and natural environment protection in industrial production, the green chemical industry is a necessary trend of development. Therefore, it is important to provide a method for preparing 7-aminomethyl-7-deazaguanine (PreQ 1) by chemical enzyme method.
Disclosure of Invention
In view of the above, the present invention has developed a method for preparing 7-aminomethyl-7-deazaguanine (PreQ 1) by enzyme catalysis and chemical synthesis, which not only shortens the preparation route, but also greatly improves the efficiency of each conversion step, and at the same time, greatly reduces the waste discharge of the whole preparation route due to the participation of enzyme catalysis, thereby improving the safety and green index in industrial production while ensuring market competitiveness.
In order to achieve the above object, the present invention provides the following technical solutions:
the invention provides a preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1), which comprises the following steps:
step 1: preparing and obtaining 7-carboxyl-7-deazaguanine (CDG);
step 2: and (3) performing enzymolysis to prepare the 7-aminomethyl-7-deazapurine (PreQ 1).
In some embodiments of the present invention, step 1 employs starting materials comprising methyl formate, methyl chloroacetate, and 2, 4-diamino-6-hydroxypyrimidine.
In some embodiments of the invention, step 1 is specifically:
step 1-1: methyl formate and methyl chloroacetate are condensed under alkaline conditions to generate an intermediate methyl 2-chloro-3-oxopropionate (compound 1);
step 1-2: condensing the intermediate with 2, 4-diamino-6-hydroxypyrimidine in aqueous solution to form 7-methyl formate-7-deazaguanine (compound 2);
step 1-3: the ester is hydrolyzed under the weak alkaline condition to obtain 7-carboxyl-7-deazaguanine (CDG).
In some embodiments of the invention, the molar ratio of methyl formate, methyl chloroacetate to 2, 4-diamino-6-hydroxypyrimidine is: (10-12): (1.5-2.5): (0.9-1.2).
Preferably, the molar ratio of methyl formate, methyl chloroacetate to 2, 4-diamino-6-hydroxypyrimidine is: 35:6:3.
In some embodiments of the invention, the temperature of the condensation in step 1-1 is from-10 to 10 ℃ for a period of from 1.5 to 4.0 hours;
the temperature of the condensation in the step 1-2 is 80-100 ℃ and the time is 0.5-2.0 h;
the temperature of the ester hydrolysis in the step 1-3 is 90-120 ℃ and the time is 2-4 h.
In some embodiments of the invention, the alkaline conditions in step 1-1 are provided by sodium methoxide;
the alkalescent condition in the step 1-3 is that the pH value is 12-14.
In some embodiments of the invention, the 7-cyano-7-deazaguanine synthetase has an enzyme activity of 75 to 120U/mg and the cyano reductase has an enzyme activity of 150 to 300U/mg.
In some embodiments of the invention, one or both of the polyphosphate kinase PPK or PTDH are also included.
In some embodiments of the invention, the enzymatic hydrolysis further comprises Adenosine Triphosphate (ATP) and Nicotinamide Adenine Dinucleotide Phosphate (NADPH), which are reaction raw materials.
The invention uses cheap industrial chemicals (methyl formate, methyl chloroacetate and 2, 4-diamino-6-hydroxy pyrimidine) as raw materials, firstly prepares 7-carboxyl-7-denitrogen guanine (CDG) through three steps of simple chemical conversion, and then converts 7-carboxyl into 7-aminomethyl with high efficiency and specificity by utilizing two steps of convenient enzyme reaction, thereby obtaining the final product 7-aminomethyl-7-denitrogen guanine (PreQ 1). The chemical-enzyme combination method utilizes the diversity of chemical catalysis and the selectivity of the enzyme method, thereby realizing the optimization of the preparation route of the PreQ1, shortening the route, improving the final yield and effectively improving the green index of the large-scale production.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below.
FIG. 1 shows a chemical enzymatic route of the present invention;
FIG. 2 shows the chemical route of 7-methyl formate-7-deazaguanine (Compound 2) in example 1;
FIG. 3 shows the chemical preparation route of 7-formic acid-7-deazaguanine (CDG) in example 2;
FIG. 4 shows the preparation of 7-cyano-7-deazapurine (PreQ) by CDG enzyme in example 3 0 );
FIG. 5 shows the preparation of 7-aminomethyl-7-deazapurine (PreQ 1) by the PreQ0 enzyme (QueF 1) in example 4;
FIG. 6 shows the preparation of 7-aminomethyl-7-deazapurine (PreQ 1) by the PreQ0 enzyme (QueF 2) in example 5;
FIG. 7 shows the direct preparation of 7-aminomethyl-7-deazapurine (PreQ 1) (QueC+QueF1) by the CDG enzyme of example 6.
Detailed Description
The invention discloses a preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1), which can be realized by appropriately improving process parameters by a person skilled in the art based on the content of the present disclosure. It is expressly noted that all such similar substitutions and modifications will be apparent to those skilled in the art, and are deemed to be included in the present invention. While the methods and applications of this invention have been described in terms of preferred embodiments, it will be apparent to those skilled in the relevant art that variations and modifications can be made in the methods and applications described herein, and in the practice and application of the techniques of this invention, without departing from the spirit or scope of the invention.
Enzyme related information:
7-cyano-7-deazaguanine synthetase (QueC synthase) derived from E.coli (Uniprot ID: P77756, EC 6.3.4.20);
cyano reductase (Nitrile Reductase, queF 1) from pectobacterium carotovorum (Pectobacterium carotovorum, uniprot ID: C6DAH4, EC 1.7.1.13);
cyano reductase (Nitrile Reductase, queF 2) from Bacillus subtilis (Bacillus subtilis, uniprot ID: O31678, EC 1.7.1.13);
polyphosphate Kinase (PPK) from Rhizobium meliloti (Rhizobium meliloti, uniprot ID: Q92SA6, EC 2.7.4.1);
the phosphite oxidase (PTDH) was engineered using a phosphite dehydrogenase in Pseudomonas stutzeri (Pseudomonas stutzeri) as a template (Uniprot ID: O69054, EC 1.20.1.1).
Fermentation production of enzyme:
the enzyme required by the invention is prepared by constructing a specific expression plasmid after the company synthesizes corresponding genes and then fermenting and producing the specific expression plasmid by escherichia coli; the method specifically comprises the following steps: after sequence optimization, the genes corresponding to the enzymes are synthesized by general biological company (Chuzhou Anhui, anhui), ndeI/XhoI restriction sites are introduced and subcloned into pET 28a expression vectors. Plasmid with correct sequence was confirmed to be transferred into E.coli (BL 21) competent cells plate culture (of the species Prinsepia) and monoclonal miniculture, the bacteria with correct protein expression are finally amplified and cultured step by step. Specifically, the single colony is transferred into 5ml LB culture solution (37 ℃) containing 50 mu M kanamycin for culture, and when the cell grows to the logarithmic phase, the cell is inoculated into 250ml LB culture solution containing the same antibiotics, and when the cell grows to the logarithmic phase, the cell is transferred into a 5L culture fermentation tank for culture, and the final protein expression is carried out. In 5L fermenter culture, 0.5mM isopropyl-beta-D-thiogalactopyranoside (IPTG) is added at 25 ℃ to induce protein expression for 6 hours when the cells OD-20, and finally, 40-70g of wet cells with over-expressed enzyme are obtained by collecting the cells (4000 rpm,20 min) by high-speed centrifugation. A small amount of cells are firstly mixed with a buffer solution (50 mM, pH 8.0) of tris (hydroxymethyl) aminomethane hydrochloride (Tris.HCl) on an ice basin uniformly, then the cells are broken by a freeze thawing method, and clear liquid is subjected to SDS-PAGE gel electrophoresis (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) after cell walls are removed by high-speed centrifugation to determine protein expression. Cells with correct protein expression were used for the next catalytic experiment, specifically, the remaining cells were mixed with Tris.HCl buffer (50 mM, pH 8.0) at low temperature (200 ml buffer mixing with 10g wet cells), then crushed cell walls at low temperature Gao Yapo, and the cell walls were removed by high speed centrifugation (16000 rpm,45 min) to obtain enzyme-containing supernatant (the enzyme activity obtained was 150-300U/ml, U was the amount of enzyme required for converting 1. Mu. Mol of substrate in one minute at room temperature). LB medium consisted of: 1% tryptone, 0.5% yeast powder, 1% NaCl,1% dipotassium hydrogen phosphate and 5% glycerol.
TABLE 1
TABLE 2
The chemical enzyme method of the invention has the route shown in figure 1:
firstly, methyl formate and methyl chloroacetate are condensed under alkaline condition to generate 2-chloro-3-oxopropionate (compound 1), then the intermediate is condensed with 2, 4-diamino-6-hydroxypyrimidine in aqueous solution to form 7-methyl formate-7-deazaguanine (compound 2), and under alkaline condition, 7-carboxyl-7-deazaguanine (CDG) is obtained by ester hydrolysis; finally, 7-carboxyl-7-deazaguanine is catalyzed by 7-cyano-7-deazaguanine synthetase (EC 6.3.4.20) to generate 7-cyano-7-deazaguanine (Pre Q0), and the Pre Q0 is prepared into a final compound 7-aminomethyl-7-deazaguanine (PreQ 1) under the action of specific cyano reductase (Nitrile Reductase, queF, EC 1.7.1.13). Since Adenosine Triphosphate (ATP) and Nicotinamide Adenine Dinucleotide Phosphate (NADPH) are required for the last two-step enzyme reaction, timely regeneration of the two coenzymes can further reduce the production cost and improve the product quality.
With public emphasis on personal safety and natural environment protection in industrial production, the green chemical industry is a necessary trend of development. Considering that a single preparation method cannot achieve efficient preparation of 7-aminomethyl-7-deazapurine, the unique advantages of both transformations by combining the use of chemo-enzymes are an important option; meanwhile, due to the environment compatibility of enzyme catalysis, waste generation in the preparation process can be greatly reduced. The preparation method combines and utilizes the preQ1 chemistry and enzyme preparation process, and forms an optimal preparation route through effective optimization and integration, and the route not only greatly reduces the production cost, but also is more suitable for large-scale industrial catalysis.
In the preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1), the raw materials and reagents used in the preparation method can be purchased from the market.
The invention is further illustrated by the following examples:
example 1: chemical preparation of 7-methyl formate-7-deazaguanine (Compound 2)
The roadmap is shown in fig. 2.
21.2g (350 mmol) of methyl formate are dissolved in 200ml of anhydrous Tetrahydrofuran (THF) and then cooled with stirring under ice bath; 4.32g (80 mmol) of sodium methoxide was added in portions at 4℃and stirred for 30 minutes after the addition was completed, followed by slow dropwise addition of 6.52g (60 mmol) of methyl chloroacetate using a dropping funnel. After the completion of the dropwise addition, the mixture was stirred at 4℃for 2 hours, and then the mixture was slowly heated to 25℃and stirred for 1 hour. And finally adding ice water into the reaction system to quench unreacted sodium methoxide, decompressing and pumping out the low-boiling point unreacted raw material methyl formate and solvent THF from the mixed solution by using a rotary evaporator, wherein the rest aqueous solution is the aqueous solution of the product methyl 2-chloro-3-oxopropionate (compound 1), and the compound can be directly subjected to the next conversion without separation. The specific procedure was to heat the aqueous solution to 90℃and then add 3.6g (30 mmol) of 2, 4-diamino-6-hydroxypyrimidine thereto, heat and maintain 90℃under stirring for 30 minutes, and then to filter and dry the solid formed in the solution to give 5.55g of 7-methyl formate-7-deazaguanine (compound 2) as a white solid (total yield 89%).
Example 2: chemical preparation of 7-methanoic acid-7-deazaguanine (CDG)
The roadmap is shown in fig. 3.
10g of 7-carboxylic acid-7-deazaguanine (48 mmol) are mixed together in a mixture of H 2 To a mixed solution of O (500 ml) and DMSO (100 ml), 6ml (5N) of an aqueous sodium hydroxide solution was then added, and the mixed solution was allowed to flow at 100℃for 2 hours. Finally, the solution was cooled to 25 ℃ during which a white solid precipitated, and the solid was filtered off and pumped off (8.5 g,91% yield), which was the target compound 7-formic acid-7-deazaguanine (CDG).
Example 3: preparation of 7-cyano-7-deazapurine from CDG enzyme (PreQ 0 )
The roadmap is shown in fig. 4.
To 500ml of a solution containing 100mM Tris-HCl pH 8.0 were added 9.7g of 7-methanoic acid-7-deazaguanine (CDG, 100 mM), 3.2g of amine chloride (120 mM), 1.4g of adenosine disodium salt ATP (5 mM), 8.4g of sodium metaphosphate (27.5 mM), 0.48g of magnesium chloride (10 mM), and 0.75g of potassium chloride (20 mM); after the pH value is adjusted to 8.0, 1000U 7-cyano-7-deazaguanine synthetase QueC (8.3-13.3 mg) and 1500U polyphosphate kinase PPK (5.0-7.5 mg) are added into the solution to start the reaction, the pH value of the reaction system is maintained to be 6.5-9.0 by adding 0.1N HCl or NaOH aqueous solution in the reaction process, the reaction is completed after stirring for 5 hours at room temperature, then HCl is added to acidify and precipitate the reactive enzyme (the pH value of the solution is adjusted to 1.5 and the reaction is rapidly stirred), protein impurities are centrifugally removed, the pH value of the solution is adjusted to 7.0, reverse osmosis is utilized to remove salt, finally D201 anion exchange resin is utilized to remove phosphoric acid impurities (deionized water is used as eluent, 7-cyano-7-deazaguanine and the resin are weakly and directly outflow), and the crude product after freeze drying is crystallized through pure water: ethanol 1:3v/v, and finally 7.4g white solid is obtained (the final yield is 84%).
Example 4: preparation of 7-aminomethyl-7-deazapurine from PreQ0 enzyme (QueF 1) (PreQ 1)
The roadmap is shown in fig. 5.
To 1000ml of a solution containing 50mM Tris-HCl pH 8.0 was added 17.5g of 7-cyano-7-deazaguanurine (100 mM), 1.5g of beta-Nicotinamide Adenine Dinucleotide Phosphate (NADP) + ) Monosodium salt (0.2 mM), 52g sodium phosphite pentahydrate (240 mM) and 200ml isopropanol. After adjusting the pH of the reaction solution to 8.0, 2000U of QueF1 and 4000U of PTDH (27-67 mg) were added to start the reaction, the reaction solution was slowly stirred at room temperature for 4 hours, and the pH of the system was maintained between 7.0 and 8.5 by adding HCl or NaOH aqueous solution during the reaction. After the reaction, HCl solution was added to precipitate proteins and the resulting solution was removed by centrifugation at high speed, and finally, the resulting solution was concentrated by desalting with a reverse osmosis membrane and crystallized with ethanol/water to obtain 15.6g of a white solid (yield: 89%).
Example 5: preparation of 7-aminomethyl-7-deazapurine from PreQ0 enzyme (QueF 2) (PreQ 1)
The roadmap is shown in fig. 6.
Similar to example 4 above, this time a QueF2 enzyme was used instead of a QueF1 enzyme. Likewise, 4.37g of 7-cyano-7-deazaguanurine (50 mM), 0.75g of β -nicotinoyl are added successively to 500ml of a solution containing 50mM of tris (hydroxymethyl) aminomethane hydrochloride (Tris. HCl) pH 8.0Amine adenine dinucleotide phosphate (NADP) + ) Monosodium salt (0.2 mM), 13g sodium phosphite pentahydrate (120 mM) and 100ml isopropanol. After adjusting the pH of the reaction solution to 8.0, 3000U of QueF2 (10-20 mg) and 4000U of PTDH are added to start the reaction (PTDH 27-67 mg), the reaction solution is slowly stirred at room temperature for 8 hours, and the pH of the system is maintained between 7.0 and 8.5 by adding HCl or NaOH aqueous solution during the reaction. After the reaction, HCl solution was added to precipitate the protein, followed by high-speed centrifugation to remove the protein, and finally, the solution was concentrated by desalting with a reverse osmosis membrane, and then, crystals were formed with ethanol/water to obtain 2.7g of a white solid (yield: 61%).
Example 6: preparation of 7-aminomethyl-7-deazapurine (PreQ 1) from CDG direct enzyme (QueC+QueF1)
The roadmap is shown in fig. 7.
To 500ml of a solution containing 100mM Tris-HCl (pH 8.0) were added 4.9g of 7-methanoic acid-7-deazaguanine (CDG, 50 mM), 1.6g of amine chloride (60 mM), 0.7g of adenosine disodium salt ATP (2.5 mM), 4.2g of sodium metaphosphate (13.8 mM), 0.48g of magnesium chloride (10 mM), 0.75g of potassium chloride (20 mM), 0.75g of beta-Nicotinamide Adenine Dinucleotide Phosphate (NADP) + ) Monosodium salt (0.2 mM), 13g sodium phosphite pentahydrate (60 mM) and 100ml isopropanol; after the pH value of the solution is adjusted to 8.0, 1000U of 7-cyano-7-deazaguanine synthetase QueC (8.3-13.3 mg), 1000U of QueF1 (3.3-6.7 mg), 2000U of polyphosphate kinase PPK (6.7-10 mg) and 3000U of PTDH (20-50 mg) are added into the solution to start the reaction, the reaction solution is slowly stirred for 6 hours at room temperature, and meanwhile, HCl or NaOH dilute aqueous solution is added at intervals in the reaction process to maintain the pH value of the system between 7.0 and 8.5. As the post-treatment of the enzyme reaction is the same, protein is precipitated by adding a dilute hydrochloric acid aqueous solution after the reaction is finished and is removed by high-speed centrifugation, a reverse osmosis membrane is utilized for desalting after the pH value of the solution is adjusted back to be neutral, D201 anion exchange resin is utilized for removing phosphoric acid impurities, and finally the obtained 7-aminomethyl-7-deazapurine crude aqueous solution is concentrated and then is crystallized by ethanol/water to obtain 4.1g of white solid (the yield of two steps is 91%).
The foregoing is merely a preferred embodiment of the present invention and it should be noted that modifications and adaptations to those skilled in the art may be made without departing from the principles of the present invention, which are intended to be comprehended within the scope of the present invention.
Sequence listing
<110> Shenzhen City Redren Biotechnology Co., ltd
<120> preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)
<130> S21P002357
<160> 10
<170> SIPOSequenceListing 1.0
<210> 1
<211> 231
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 1
Met Lys Arg Ala Val Val Val Phe Ser Gly Gly Gln Asp Ser Thr Thr
1 5 10 15
Cys Leu Val Gln Ala Leu Gln Gln Tyr Asp Glu Val His Cys Val Thr
20 25 30
Phe Asp Tyr Gly Gln Arg His Arg Ala Glu Ile Asp Val Ala Arg Glu
35 40 45
Leu Ala Leu Lys Leu Gly Ala Arg Ala His Lys Val Leu Asp Val Thr
50 55 60
Leu Leu Asn Glu Leu Ala Val Ser Ser Leu Thr Arg Asp Ser Ile Pro
65 70 75 80
Val Pro Asp Tyr Glu Pro Glu Ala Asp Gly Ile Pro Asn Thr Phe Val
85 90 95
Pro Gly Arg Asn Ile Leu Phe Leu Thr Leu Ala Ala Ile Tyr Ala Tyr
100 105 110
Gln Val Lys Ala Glu Ala Val Ile Thr Gly Val Cys Glu Thr Asp Phe
115 120 125
Ser Gly Tyr Pro Asp Cys Arg Asp Glu Phe Val Lys Ala Leu Asn His
130 135 140
Ala Val Ser Leu Gly Met Ala Lys Asp Ile Arg Phe Glu Thr Pro Leu
145 150 155 160
Met Trp Ile Asp Lys Ala Glu Thr Trp Ala Leu Ala Asp Tyr Tyr Gly
165 170 175
Lys Leu Asp Leu Val Arg Asn Glu Thr Leu Thr Cys Tyr Asn Gly Phe
180 185 190
Lys Gly Asp Gly Cys Gly His Cys Ala Ala Cys Asn Leu Arg Ala Asn
195 200 205
Gly Leu Asn His Tyr Leu Ala Asp Lys Pro Thr Val Met Ala Ala Met
210 215 220
Lys Gln Lys Thr Gly Leu Arg
225 230
<210> 2
<211> 282
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 2
Met Ser Val Tyr Asp Lys His Gln Ala Leu Ser Gly Leu Thr Leu Gly
1 5 10 15
Lys Pro Thr Pro Tyr His Asp Arg Tyr Asp Ala Ala Leu Leu Gln Pro
20 25 30
Val Pro Arg Ser Leu Asn Arg Asp Pro Leu Gly Ile His Pro Asp Ser
35 40 45
Leu Pro Phe His Gly Ala Asp Ile Trp Thr Leu Tyr Glu Leu Ser Trp
50 55 60
Leu Asn Asn Arg Gly Val Pro Gln Val Ala Val Gly Glu Met His Leu
65 70 75 80
Asn Ala Glu Ser Leu Asn Leu Ile Glu Ser Lys Ser Phe Lys Leu Tyr
85 90 95
Leu Asn Ser Phe Asn Gln Thr Thr Phe Asp Ser Trp Glu Ser Val Arg
100 105 110
Ala Thr Leu Ala Asn Asp Leu Ala His Cys Ala Gln Gly Asp Val Ser
115 120 125
Ile Thr Leu Phe Lys Leu Ser Glu Leu Glu Gly Gln Pro Leu Ala Gly
130 135 140
Phe Thr Gly Glu Cys Ile Asp Asp Gln Asp Ile Gln Ile Asp Ser Tyr
145 150 155 160
Asp Phe Asn Ala Asp Tyr Leu Ala Thr Asn Glu Gln Asp Ala Pro Val
165 170 175
Val Glu Glu Thr Leu Val Ser His Leu Leu Lys Ser Asn Cys Leu Ile
180 185 190
Thr His Gln Pro Asp Trp Gly Ser Val Gln Ile His Tyr Arg Gly Lys
195 200 205
Arg Ile Asn Arg Glu Ala Leu Leu Arg Tyr Ile Ile Ser Phe Arg His
210 215 220
His Asn Glu Phe His Glu Gln Cys Val Glu Arg Ile Phe Asn Asp Ile
225 230 235 240
Met Arg Tyr Tyr Gln Pro Glu Lys Leu Ser Val Tyr Ala Arg Tyr Thr
245 250 255
Arg Arg Gly Gly Leu Asp Ile Asn Pro Trp Arg Ser Asn Thr Ala Phe
260 265 270
Asn Ala Pro Asn Gly Arg Leu Pro Arg Gln
275 280
<210> 3
<211> 165
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 3
Met Thr Thr Arg Lys Glu Ser Glu Leu Glu Gly Val Thr Leu Leu Gly
1 5 10 15
Asn Gln Gly Thr Asn Tyr Leu Phe Glu Tyr Ala Pro Asp Val Leu Glu
20 25 30
Ser Phe Pro Asn Lys His Val Asn Arg Asp Tyr Phe Val Lys Phe Asn
35 40 45
Cys Pro Glu Phe Thr Ser Leu Cys Pro Lys Thr Gly Gln Pro Asp Phe
50 55 60
Ala Thr Ile Tyr Ile Ser Tyr Ile Pro Asp Glu Lys Met Val Glu Ser
65 70 75 80
Lys Ser Leu Lys Leu Tyr Leu Phe Ser Phe Arg Asn His Gly Asp Phe
85 90 95
His Glu Asp Cys Met Asn Ile Ile Met Asn Asp Leu Ile Glu Leu Met
100 105 110
Asp Pro Arg Tyr Ile Glu Val Trp Gly Lys Phe Thr Pro Arg Gly Gly
115 120 125
Ile Ser Ile Asp Pro Tyr Thr Asn Tyr Gly Lys Pro Gly Thr Lys Tyr
130 135 140
Glu Lys Met Ala Glu Tyr Arg Met Met Asn His Asp Leu Tyr Pro Glu
145 150 155 160
Thr Ile Asp Asn Arg
165
<210> 4
<211> 300
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 4
Met Ala Leu Asp Glu Ala Pro Ala Glu Ala Arg Pro Gly Ser Arg Ala
1 5 10 15
Val Glu Leu Glu Ile Asp Gly Arg Ser Arg Ile Phe Asp Ile Asp Asp
20 25 30
Pro Asp Leu Pro Lys Trp Ile Asp Glu Glu Ala Phe Arg Ser Asp Asp
35 40 45
Tyr Pro Tyr Lys Lys Lys Leu Asp Arg Glu Glu Tyr Glu Glu Thr Leu
50 55 60
Thr Lys Leu Gln Ile Glu Leu Val Lys Val Gln Phe Trp Met Gln Ala
65 70 75 80
Thr Gly Lys Arg Val Met Ala Val Phe Glu Gly Arg Asp Ala Ala Gly
85 90 95
Lys Gly Gly Ala Ile His Ala Thr Thr Ala Asn Met Asn Pro Arg Ser
100 105 110
Ala Arg Val Val Ala Leu Thr Lys Pro Thr Glu Thr Glu Arg Gly Gln
115 120 125
Trp Tyr Phe Gln Arg Tyr Val Ala Thr Phe Pro Thr Ala Gly Glu Phe
130 135 140
Val Leu Phe Asp Arg Ser Trp Tyr Asn Arg Ala Gly Val Glu Pro Val
145 150 155 160
Met Gly Phe Cys Thr Pro Asp Gln Tyr Glu Gln Phe Leu Lys Glu Ala
165 170 175
Pro Arg Phe Glu Glu Met Ile Ala Asn Glu Gly Ile His Leu Phe Lys
180 185 190
Phe Trp Ile Asn Ile Gly Arg Glu Met Gln Leu Lys Arg Phe His Asp
195 200 205
Arg Arg His Asp Pro Leu Lys Ile Trp Lys Leu Ser Pro Met Asp Ile
210 215 220
Ala Ala Leu Ser Lys Trp Asp Asp Tyr Thr Gly Lys Arg Asp Arg Met
225 230 235 240
Leu Lys Glu Thr His Thr Glu His Gly Pro Trp Ala Val Ile Arg Gly
245 250 255
Asn Asp Lys Arg Arg Ser Arg Ile Asn Val Ile Arg His Met Leu Thr
260 265 270
Lys Leu Asp Tyr Asp Gly Lys Asp Glu Ala Ala Ile Gly Glu Val Asp
275 280 285
Glu Lys Ile Leu Gly Ser Gly Pro Gly Phe Leu Arg
290 295 300
<210> 5
<211> 336
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 5
Met Leu Pro Lys Leu Val Ile Thr His Arg Val His Asp Glu Ile Leu
1 5 10 15
Gln Leu Leu Ala Pro His Cys Glu Leu Met Thr Asn Gln Thr Asp Ser
20 25 30
Thr Leu Thr Arg Glu Glu Ile Leu Arg Arg Cys Arg Asp Ala Gln Ala
35 40 45
Met Met Ala Phe Met Pro Asp Arg Val Asp Ala Asp Phe Leu Gln Ala
50 55 60
Cys Pro Glu Leu Arg Val Ile Gly Cys Ala Leu Lys Gly Phe Asp Asn
65 70 75 80
Phe Asp Val Asp Ala Cys Thr Ala Arg Gly Val Trp Leu Thr Phe Val
85 90 95
Pro Asp Leu Leu Thr Val Pro Thr Ala Glu Leu Ala Ile Gly Leu Ala
100 105 110
Val Gly Leu Gly Arg His Leu Arg Ala Ala Asp Ala Phe Val Arg Ser
115 120 125
Gly Lys Phe Arg Gly Trp Gln Pro Arg Phe Tyr Gly Thr Gly Leu Asp
130 135 140
Asn Ala Thr Val Gly Phe Leu Gly Met Gly Ala Ile Gly Leu Ala Met
145 150 155 160
Ala Asp Arg Leu Gln Gly Trp Gly Ala Thr Leu Gln Tyr His Glu Arg
165 170 175
Lys Ala Leu Asp Thr Gln Thr Glu Gln Arg Leu Gly Leu Arg Gln Val
180 185 190
Ala Cys Ser Glu Leu Phe Ala Ser Ser Asp Phe Ile Leu Leu Ala Leu
195 200 205
Pro Leu Asn Ala Asp Thr Leu His Leu Val Asn Ala Glu Leu Leu Ala
210 215 220
Leu Val Arg Pro Gly Ala Leu Leu Val Asn Pro Cys Arg Gly Ser Val
225 230 235 240
Val Asp Glu Ala Ala Val Leu Ala Ala Leu Glu Arg Gly Gln Leu Gly
245 250 255
Gly Tyr Ala Ala Asp Val Phe Glu Met Glu Asp Trp Ala Arg Ala Asp
260 265 270
Arg Pro Gln Gln Ile Asp Pro Ala Leu Leu Ala His Pro Asn Thr Leu
275 280 285
Phe Thr Pro His Ile Gly Ser Ala Val Arg Ala Val Arg Leu Glu Ile
290 295 300
Glu Arg Cys Ala Ala Gln Asn Ile Leu Gln Ala Leu Ala Gly Glu Arg
305 310 315 320
Pro Ile Asn Ala Val Asn Arg Leu Pro Lys Ala Glu Pro Ala Ala Cys
325 330 335
<210> 6
<211> 696
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 6
atgaaacgtg ctgtcgttgt gttcagtgga ggtcaggatt ccaccacctg tctggtgcag 60
gcattacaac aatatgatga agtccattgc gtgacgttcg attacggtca gcggcatcgc 120
gcagaaatcg acgtggcacg cgaactggcg ctgaaactgg gggcacgcgc gcataaggtg 180
ctggatgtca ccctgctcaa cgagctggcg gtcagtagcc tgacgcgtga cagcattccg 240
gtgcctgatt atgaacctga agccgatggt atcccgaata cgtttgtccc agggcgtaat 300
attttgttcc tgacgctggc ggcaatatat gcgtatcagg taaaagcaga agccgtaatt 360
actggcgtct gcgaaacgga tttctccggc tacccggatt gccgcgatga gtttgtgaaa 420
gcactaaacc atgccgtcag tttgggcatg gcgaaagata ttcgttttga aacgccgctg 480
atgtggattg ataaagcgga aacctgggcg ctggcagatt attacggcaa actggattta 540
gtccgtaacg aaacgttgac ctgctataac ggctttaaag gcgacggttg cggtcattgt 600
gcggcatgta atttacgcgc caacggtttg aatcattatc tggccgataa accgacggtg 660
atggcagcga tgaagcagaa aaccgggttg aggtaa 696
<210> 7
<211> 849
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 7
atgtccgttt atgacaagca ccaggccctg agcgggctga cattgggcaa acccactccc 60
taccacgacc gctatgatgc cgcccttctg caacccgtgc cacgtagcct gaaccgcgat 120
ccactcggca ttcatcctga tagcctacct tttcatggcg cagatatctg gacgctctac 180
gagctttcct ggctgaacaa ccgtggcgtg cctcaggtag ccgtcggtga aatgcatctc 240
aatgcggaaa gcctgaatct gattgaatca aaaagtttta agctgtacct gaacagcttt 300
aatcagacga cattcgacag ttgggagagc gtacgcgcga cgttagccaa cgacctggcg 360
cactgtgcac agggggacgt cagcatcacg cttttcaaac tcagcgagct cgaaggccag 420
ccgctagcgg gattcactgg cgaatgcatc gacgatcaag acattcagat cgacagctac 480
gacttcaacg ccgactatct ggcgacaaac gaacaggacg cgcctgtcgt tgaagaaacg 540
ctggtcagcc acctgctgaa atccaactgt ttgatcaccc atcagcccga ctggggctct 600
gtacagatcc actatcgcgg caaacgcatt aaccgtgaag cactgctgcg ctacattatc 660
tcgtttcgtc atcataacga atttcatgaa cagtgtgtgg aacgaatttt taacgacatc 720
atgcgctact accagccgga aaaactcagc gtttacgccc gctatacccg acgcggcggg 780
ctggacatca acccgtggcg cagcaatacc gcgtttaacg caccaaatgg acgcctgccg 840
cgtcagtaa 849
<210> 8
<211> 498
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 8
atgacgacaa gaaaagaatc agaattagaa ggtgtaacat tgctaggcaa tcaaggtaca 60
aattatttgt tcgaatatgc accggacgtg ctggaatcct tccctaataa acatgtaaac 120
cgtgattact ttgtaaaatt caattgcccg gaattcacat ctttatgtcc taaaacaggc 180
cagcctgact ttgcgacaat ctacatcagc tacattcctg atgaaaaaat ggttgaaagc 240
aaatcattaa agctgtatct attcagcttc agaaaccatg gtgacttcca cgaggactgc 300
atgaatatca tcatgaacga cttgattgaa ttaatggacc cgcgctacat tgaagtatgg 360
ggcaaattca cgccaagagg cggaatttcc attgatccgt acacaaacta cggaaagcct 420
ggcacgaagt atgagaaaat ggccgaatac cgtatgatga accatgattt gtatccggag 480
acaattgata atcgttaa 498
<210> 9
<211> 903
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 9
atggcactcg acgaagcacc ggccgaagca aggccgggga gccgggcggt cgaactggag 60
atcgacggca gaagccgcat cttcgacatc gacgatccgg acctgccgaa atggatcgac 120
gaggaggcct tccgctccga cgattacccc tacaagaaaa aactcgatcg ggaggaatac 180
gaagaaacgc tgacgaagct gcagatcgaa ctggtcaagg tccagttctg gatgcaggcg 240
accggcaagc gcgtgatggc ggtcttcgag ggacgcgacg ctgccggcaa gggtggtgcg 300
atccacgcga cgacggccaa tatgaacccc cgctccgcgc gcgtcgtcgc actgacgaaa 360
ccgacggaga ccgaacgggg ccagtggtac ttccagcgct atgtcgcaac cttcccgacc 420
gccggcgagt tcgtcctttt cgaccgctcc tggtacaacc gcgccggtgt cgaaccggtc 480
atgggctttt gcacccccga ccagtacgag caattcctta aagaggcgcc ccgcttcgag 540
gagatgatcg cgaacgaggg catccatctc ttcaagtttt ggatcaatat cggccgggaa 600
atgcaactga agcgcttcca tgaccggcgc cacgatccgt tgaagatctg gaagctttcg 660
ccgatggaca tcgcggcgct gagcaagtgg gacgactaca ccggaaaacg cgaccgtatg 720
ctgaaggaaa cgcacacgga gcacgggcca tgggcggtca tccgcggcaa cgacaagcgc 780
cgctcgcgga tcaacgtgat ccgccacatg ctgacgaagc tcgactatga cggcaaggac 840
gaggcggcga tcggagaggt cgacgaaaag atcctcggct ccggccccgg ttttctcagg 900
tga 903
<210> 10
<211> 1011
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 10
atgttaccga aattagttat cacgcacaga gtgcacgacg aaatccttca attgctggcc 60
cctcattgtg agttgatgac caaccaaacc gattctaccc tgacgagaga agagatactg 120
cgccgttgca gagacgcaca agccatgatg gcgtttatgc cggaccgtgt agatgcagac 180
tttcttcaag cttgcccgga acttcgggtc attggttgtg ctttgaaagg gttcgacaac 240
tttgacgtgg atgcgtgtac tgcacgcggg gtatggctta cttttgtacc tgacttattg 300
acggttccca ctgccgagct tgctattggc ctggccgtcg gattaggccg ccatttacgt 360
gcggcagatg cgttcgtacg gagtgggaag tttcggggct ggcaaccgcg attctacggg 420
actggattgg ataacgccac tgtaggtttc cttgggatgg gtgccatagg tttagctatg 480
gcagatagat tacaggggtg gggagctacc cttcaatatc atgagcgtaa agcattggat 540
acacaaacag aacagcgctt gggtcttaga caggtcgcgt gctcggaact tttcgcttcc 600
tcagacttca tactgttggc cttgccactt aacgctgaca ctctacattt ggtaaacgct 660
gaattgctgg ctttggtacg tcccggcgca ctgttagtta atccgtgccg gggctcggtg 720
gtagacgagg cagccgtgct ggcagcgctt gagagagggc aacttggcgg atatgctgca 780
gacgtgttcg agatggaaga ctgggcccgc gcggaccgtc cacagcaaat cgatcctgcg 840
ttgttggccc accctaatac tttatttact ccgcacatcg gatcagcggt gagagcggtg 900
cggcttgaga ttgagcgttg cgcagctcag aacatcctcc aggcgctggc aggagaacgt 960
ccaattaatg ctgtaaatcg tttaccgaag gctgaaccag cagcttgttg a 1011

Claims (5)

  1. The preparation method of the 7-aminomethyl-7-deazaguanine is characterized by comprising the following steps:
    step 1: preparing 7-formic acid-7-deazaguanine;
    step 2: the 7-aminomethyl-7-deazaguanine is prepared through enzymolysis;
    the raw materials adopted in the step 1 comprise methyl formate, methyl chloroacetate and 2, 4-diamino-6-hydroxypyrimidine;
    the step 1 specifically comprises the following steps:
    step 1-1: methyl formate and methyl chloroacetate are condensed under alkaline conditions to generate an intermediate methyl 2-chloro-3-oxopropionate;
    step 1-2: condensing the intermediate with 2, 4-diamino-6-hydroxypyrimidine in aqueous solution to form 7-methyl formate-7-deazaguanine;
    step 1-3: ester hydrolysis under weak alkaline condition to obtain 7-formic acid-7-deazaguanine;
    the enzymes in the step 2 are 7-cyano-7-deazaguanine synthetase, cyano reductase, polyphosphate kinase and phosphite oxidase;
    the reaction raw materials for enzymolysis are adenosine triphosphate and nicotinamide adenine dinucleotide phosphate;
    the amino acid sequence of the 7-cyano-7-deazaguanine synthetase is shown as SEQ ID No. 1;
    the amino acid sequences of the cyano reductase are shown as SEQ ID No.2 and SEQ ID No. 3;
    the amino acid sequence of the polyphosphate kinase is shown as SEQ ID No. 4;
    the amino acid sequence of the phosphite oxidase is shown as SEQ ID No. 5.
  2. 2. The process according to claim 1, wherein the molar ratio of methyl formate, methyl chloroacetate to 2, 4-diamino-6-hydroxypyrimidine is: (10-12): (1.5-2.5): (0.9-1.2).
  3. 3. The method according to claim 2, wherein the temperature of the condensation in step 1-1 is-10 to 10 ℃ for 1.5 to 4.0 hours;
    the temperature of the condensation in the step 1-2 is 80-100 ℃ and the time is 0.5-2 h;
    the temperature of the ester hydrolysis in the step 1-3 is 90-120 ℃ and the time is 2-4 h.
  4. 4. A process according to claim 3, wherein the alkaline conditions in step 1-1 are provided by sodium methoxide;
    the alkalescent condition in the step 1-3 is that the pH value is 12-14.
  5. 5. The process according to claim 4, wherein the enzyme activity of the 7-cyano-7-deazaguanine synthase is 75 to 120U/mg and the enzyme activity of the cyano-reductase is 150 to 300U/mg.
CN202111166689.1A 2021-09-30 2021-09-30 Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1) Active CN113881728B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111166689.1A CN113881728B (en) 2021-09-30 2021-09-30 Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111166689.1A CN113881728B (en) 2021-09-30 2021-09-30 Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)

Publications (2)

Publication Number Publication Date
CN113881728A CN113881728A (en) 2022-01-04
CN113881728B true CN113881728B (en) 2023-12-15

Family

ID=79005297

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111166689.1A Active CN113881728B (en) 2021-09-30 2021-09-30 Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)

Country Status (1)

Country Link
CN (1) CN113881728B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015697A (en) * 1996-11-21 2000-01-18 Ajinomoto Co., Inc. Method for producing nucleoside-5'-phosphate ester
US7364882B1 (en) * 2004-09-24 2008-04-29 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Portland State University Enzymatic reduction of a nitrile containing compound to the corresponding amine
WO2013041969A2 (en) * 2011-09-21 2013-03-28 King Abdullah University Of Science And Technology Didemnin biosynthetic gene cluster in tistrella mobilis
WO2020185775A2 (en) * 2019-03-11 2020-09-17 University Of Florida Research Foundation, Inc. Materials and methods for reducing nucleic acid degradation in bacteria
CN113234698A (en) * 2021-05-07 2021-08-10 深圳瑞德林生物技术有限公司 Preparation method of cyano reductase and gabapentin

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008141119A2 (en) * 2007-05-09 2008-11-20 Vertex Pharmaceuticals Incorporated Modulators of cftr

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015697A (en) * 1996-11-21 2000-01-18 Ajinomoto Co., Inc. Method for producing nucleoside-5'-phosphate ester
US7364882B1 (en) * 2004-09-24 2008-04-29 State Of Oregon Acting By And Through The State Board Of Higher Education On Behalf Of Portland State University Enzymatic reduction of a nitrile containing compound to the corresponding amine
WO2013041969A2 (en) * 2011-09-21 2013-03-28 King Abdullah University Of Science And Technology Didemnin biosynthetic gene cluster in tistrella mobilis
WO2020185775A2 (en) * 2019-03-11 2020-09-17 University Of Florida Research Foundation, Inc. Materials and methods for reducing nucleic acid degradation in bacteria
CN113234698A (en) * 2021-05-07 2021-08-10 深圳瑞德林生物技术有限公司 Preparation method of cyano reductase and gabapentin

Also Published As

Publication number Publication date
CN113881728A (en) 2022-01-04

Similar Documents

Publication Publication Date Title
CN112795606B (en) Enzymatic synthesis method of beta-nicotinamide mononucleotide
TWI695888B (en) A composition for producing tagatose and methods for producing tagatose using the same
CN107922952B (en) Method for preparing nicotinamide mononucleotide
CN111621482B (en) Glufosinate-ammonium dehydrogenase mutant, gene engineering bacteria and one-pot multi-enzyme synchronous directed evolution method
CN111321193B (en) Method for asymmetrically preparing L-glufosinate-ammonium by redox through biological multi-enzyme coupling method
CN111363775B (en) Method for preparing L-glufosinate-ammonium by racemization removal with biological enzyme method, glufosinate-ammonium dehydrogenase mutant and application
JP2014503184A (en) O-phosphoserine sulfhydrylase mutant and method for producing cysteine using the same
CN113151198B (en) Gamma-glutamine synthetase mutant, coding gene, amino acid sequence and application thereof
JP6731119B2 (en) Gene encoding L-alanyl-L-glutamine biosynthesis enzyme and use thereof
CN112280755B (en) Mutant enzyme, application thereof and process for preparing sanshengtai by enzyme catalysis method
CN112980906B (en) Enzyme composition for preparing beta-nicotinamide mononucleotide and application thereof
CN113151199B (en) Gamma-glutamine synthetase mutant with thermal stability, coding gene, amino acid sequence and application thereof
CN111808829B (en) Gamma-glutamyl methylamine synthetase mutant and application thereof
CN113881728B (en) Preparation method of 7-aminomethyl-7-deazaguanine (PreQ 1)
CN112813012A (en) Genetically engineered bacterium, preparation method thereof and application thereof in cysteine production
JP5209639B2 (en) Method for producing novel N-acetylglucosamine-2-epimerase and CMP-N-acetylneuraminic acid
CN114657199B (en) Recombinant engineering bacterium and application thereof in preparation of D-pantothenic acid
CN113151378B (en) Method for preparing nucleoside, nicotinic acid adenine dinucleotide and nicotinic acid mononucleotide of nicotinic acid or derivative thereof, enzyme composition and application
CN112458073B (en) H-protein mutant and application thereof
CN115537405B (en) Ketoreductase and application thereof in preparation of (S) -1- (3-chlorophenyl) -1, 3-propanediol
CN114657200B (en) Recombinant engineering bacterium and method for preparing D-pantoic acid by using same
CN110804634A (en) Process for preparing 2,4-diaminobutyric acid by enzyme catalysis method
JP2003093091A5 (en)
CN114657198B (en) Recombinant engineering bacterium and application thereof in preparation of panto-compound
CN116855473A (en) Polyphosphokinase mutant and application thereof in preparation of citicoline

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
CB02 Change of applicant information

Address after: 518000 floor 3, building 2, Shenzhen biological incubation base, No. 10, Gaoxin Zhongyi Road, Maling community, Yuehai street, Nanshan District, Shenzhen, Guangdong

Applicant after: SHENZHEN READLINE BIOTECHNOLOGY Co.,Ltd.

Address before: 518000 Room 101, 1st floor, building 2, Shenzhen biological incubation base, No.10, Gaoxin middle 1st Road, Maling community, Yuehai street, Nanshan District, Shenzhen City, Guangdong Province

Applicant before: SHENZHEN READLINE BIOTECHNOLOGY Co.,Ltd.

CB02 Change of applicant information
GR01 Patent grant
GR01 Patent grant