CN112725227B - Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II - Google Patents

Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II Download PDF

Info

Publication number
CN112725227B
CN112725227B CN202011604925.9A CN202011604925A CN112725227B CN 112725227 B CN112725227 B CN 112725227B CN 202011604925 A CN202011604925 A CN 202011604925A CN 112725227 B CN112725227 B CN 112725227B
Authority
CN
China
Prior art keywords
germicidin
streptomyces
alpha
hexokinase
compound
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
CN202011604925.9A
Other languages
Chinese (zh)
Other versions
CN112725227A (en
Inventor
潘华奇
白岩
胡江春
姚雪春
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.)
Institute of Applied Ecology of CAS
Original Assignee
Institute of Applied Ecology of CAS
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 Institute of Applied Ecology of CAS filed Critical Institute of Applied Ecology of CAS
Priority to CN202011604925.9A priority Critical patent/CN112725227B/en
Publication of CN112725227A publication Critical patent/CN112725227A/en
Application granted granted Critical
Publication of CN112725227B publication Critical patent/CN112725227B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/34Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D309/36Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms
    • C07D309/38Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with oxygen atoms directly attached to ring carbon atoms one oxygen atom in position 2 or 4, e.g. pyrones
    • 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
    • C12P17/02Oxygen as only ring hetero atoms
    • C12P17/06Oxygen as only ring hetero atoms containing a six-membered hetero ring, e.g. fluorescein
    • 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
    • C12P17/10Nitrogen as only ring hetero atom
    • 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
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • 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
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/40Preparation of oxygen-containing organic compounds containing a carboxyl group including Peroxycarboxylic acids
    • C12P7/42Hydroxy-carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to the field of microbial medicines, in particular to sponge-derived streptomyces with a hexokinase II inhibition effect and preparation and application of a hexokinase II inhibition active substance thereof. The marine Streptomyces is Streptomyces sp.18A01, is preserved in China general microbiological culture collection management center at 12.15.2020, and has a preservation number: CGMCC 21351. The strain can also be used for preparing compounds such as alpha-pyrones, ferulic acid, alpha-ribazone, N-acetyltryptophan and the like by fermentation, and particularly preparing a novel alpha-pyrone compound germicidins P-S (1-4). The prepared alpha-pyrone substance is used as an inhibitor of hexokinase II and has application prospects in development of candidate drugs of antitumor drugs and the like.

Description

Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II
Technical Field
The invention relates to the field of microbial medicines, in particular to sponge-derived streptomyces with a hexokinase II inhibition effect and preparation and application of a hexokinase II inhibition active substance thereof.
Background
Actinomycetes are widely distributed in nature and they produce metabolites with a variety of biological functions. About 50% of the natural products of microbial origin that have been found to date originate from actinomycetes, whereas streptomyces contributes about 70% of their active substances. In recent years, the number of new compounds discovered from marine streptomyces has been on an increasing trend year by year, and about 67% of natural products of marine streptomyces exhibit cytotoxic, bacteriostatic, antimalarial and antiparasitic activities (marine science, 2016,51: 86-124).
Streptomyces antifocyanicus and Streptomyces humifusus have been reported, secondary metabolic activity products in Streptomyces tricolor have not been reported, Streptomyces coelsescens can prevent and treat aphid disease (CN106967629-A), and also can produce a series of glycerolipids with stain removing effect (biosci. Biotech. Bioch.,2012,76: 1746-. However, in general, the bioactive secondary metabolites of the streptomyces group with the highest sequence homology of 16S rDNA are less reported and need to be deeply and widely excavated.
Hexokinase (Hexokinase II, HK2), an enzyme that catalyzes the phosphorylation of hexose sugars, is expressed in small amounts in normal tissues. It is the first enzyme of the glycolytic pathway and also the rate-limiting enzyme of the glycolytic pathway (nat. Commun.2018,9,446). Glycolysis often occurs when cells are hypoxic and the division of tumor cells is vigorous, requiring enormous energy. It has been reported that tumor tissue acquires energy mainly by glycolysis even under conditions where oxygen is sufficiently supplied. The increase of the expression amount and activity of hexokinase in tumor tissue ensures that the tumor tissue can still ensure enough energy under the condition of lacking oxygen, and many intermediates of glycolysis can be utilized by tumor cells to synthesize protein, nucleic acid, lipid and the like, thereby providing necessary material basis for the growth and proliferation of the tumor cells (J.Nucl.Med.2002,43, 173-. Researches find that the characteristics of high sugar metabolism of tumors are as follows: approximately 60% of the ATP in tumor cells is derived from the glycolytic pathway, whose hexokinase content has been shown to be elevated in resected lung, gastrointestinal and breast malignant tissues, and in breast cancer it has been found that hexokinase activity is higher when the lesions metastasize (Eur. J. biochem. 2000,267, 6067-6073). It has also been reported that hexokinase activity is increased in renal tumor tissue (biochem. mol. Med.1995,54: 53-58).
Therefore, if an effective hexokinase inhibitor can be found and the activity of the hexokinase inhibitor is inhibited, glycolysis of tumor cells is inhibited, so that the tumor cells cannot obtain enough glucose-6-phosphate, ATP cannot be converted into ADP to provide energy for various activities of the cells, and the tumor cells are apoptotic. The invention provides a marine streptomyces with hexokinase II inhibition effect, and a novel alpha-pyrone hexokinase inhibitor and other active products can be prepared by utilizing the marine streptomyces.
Disclosure of Invention
The invention aims to provide marine streptomyces with a hexokinase II inhibition effect and preparation and application of a hexokinase II inhibition active substance thereof.
In order to achieve the purpose, the invention adopts the technical scheme that:
a Streptomyces spongiensis, Streptomyces sp.18A01, is preserved in China general microbiological culture collection management center at 12.10.12.2020 with the preservation number: CGMCC 21351.
The marine streptomyces contains an 8.76Mbp genome; the genome contains clusters of genes that synthetically produce Desferrioxamine B, Geosmin, Isorenieratene, Hopen, Albaflavone, Germidin, SapB (lanthionide), Spore fragment, Calcium-dependent antibody (CDA), Coelimycin P1, Undeclyprodigoside, Coelicidin, Melanin, Coelictin and Actinorhoddin backbone compounds.
An application of marine streptomyces, namely an application of a metabolic product of the marine streptomyces 18A01 as a hexokinase II inhibitor or an anti-tumor candidate drug.
An application of marine streptomycete, namely an application of a metabolic product of the marine streptomycete 18A01 in preparing any one of compounds of alpha-pyrone germicidins, ferulic acid, alpha-ribazole and N-acetyltryptophan.
The preparation of the marine streptomyces 18A01 metabolite comprises the following steps:
1) inoculating the marine streptomyces 18A01 on an ISP3 solid culture medium for 2-5d activation, then inoculating on an ISP3 liquid culture medium containing artificial sea salt, and culturing at 28 ℃ for 2-4d to be used as seed liquid; inoculating the fermented seed liquid 2-10% by volume to ISP3 liquid culture medium containing artificial sea salt, and performing shaking culture at 28 deg.C for 5-10 d;
2) centrifuging the fermentation liquor to remove thallus to obtain fermentation supernatant, and adsorbing active ingredients in the fermentation liquor by using macroporous adsorption resin XAD16 solid phase, wherein the volume percentage of the resin to the fermentation liquor is 3-6%; after adsorption is completed for 2h-4h, the resin is washed by distilled water, then desorbed by methanol, and a crude extract A obtained after the methanol solvent is recovered is the metabolic product of the marine streptomyces 18A 01.
Further purifying and preparing the crude extract A to obtain germicidins, ferulic acid, alpha-ribazole and N-acetyltryptophan: separating the crude extract A by adopting a flash silica gel column chromatography, and performing separation according to a dichloromethane ratio: gradient eluting with methanol (v/v) ratio of 100:0-0:100 to obtain 4 fractions (F)A-FD). Fraction FBThe compounds germicidin P (1), germicidin Q (2), germicidin R (3), germicidin S (4), germicidin L (11), germicidin M (12) and (1S,2S) -germicidin N (13) are obtained by adopting semi-preparative reverse phase HPLC and eluting with 35% methanol aqueous solution at the flow rate of 2.5 mL/min. Fraction FARemoving impurities by gel LH20, adopting semi-preparative reverse phase HPLC, eluting by 40% -55% methanol water solution with the flow rate of 2.5mL/min to obtain compounds germicidin A (5), germicidin B (6), isogermicidin B (7), germicidin C (8), germicidin D (9), germicidin H (10) and ferulic acid (14), and mixing the fractions FCRemoving impurities by gel LH20, eluting with 25% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain compound alpha-ribavirin (15), and collecting fraction FDRemoving impurities by gel LH20, adopting semi-preparative reverse phase HPLC, eluting with 30% methanol water solution at flow rate of 2.5mL/min to obtain compound N-acetyltryptophan (16).
A new alpha-pyrone (germicidin) compound is generated from marine streptomyces, the alpha-pyrone (germicidin P, germicidin Q, germicidin R and germicidin S) compound is shown as (I),
Figure RE-GDA0002979895260000031
the application of alpha-pyrone (germicidin) compounds, wherein the compounds germicidins P (1), Q (2), R (3), S (4), D (9), H (10), L (11), M (12) and (1S,2S) -germicidin N (13) can be used as hexokinase II inhibitors.
The alpha-pyrone (germicidin) compound is applied to serving as an anti-tumor candidate drug.
Furthermore, the compound germicidin R (3) can be used as an anti-tumor candidate drug; wherein the tumor is colon cancer HCT 116.
The invention has the advantages that:
1. the obtained strain 18A01 has uniqueness, the living environment is specially from sponge in the sea area of Yongxing island, and the strain has the potential of generating various active substances. Several strains with the same 16S rDNA sequence have not been reported to be capable of producing compounds such as alpha-pyrones, ferulic acid, alpha-ribazole and N-acetyltryptophan by fermentation, and the genome ANI is lower than 18A01, showing that the sequence is obviously different from 18A 01.
2. The strain 18A01 can simultaneously prepare alpha-pyrones, ferulic acid, alpha-ribazole and N-acetyltryptophan type compounds, and is the first invention; among them, alpha-pyrone germicidins A-C has been reported to have a hexokinase II inhibitory effect, but germicidins D, H, L-N, P-S are invented for the first time as hexokinase II inhibitors.
3. Four novel alpha-pyrone germicidins P-S compounds are invented for the first time.
Drawings
FIG. 1 is a colony morphology diagram of marine Streptomyces provided by the invention.
FIG. 2 shows the taxonomic status of 18A01 for the phylogenetic tree based on the 16S rDNA sequence provided by the present invention.
FIG. 3 shows the alpha-pyrone type secondary metabolites produced by strain 18A01 in ISP3 fermentation medium.
FIG. 4 is a chemical structural diagram of compounds 1-16 obtained by using the strain 18A01 according to the present invention.
FIG. 5 provides HRESIMS spectra of Germicidin P (1) obtained using strain 18A01 according to the invention.
FIG. 6 shows Germicidins P-S (1-4) obtained by using the strain 18A01 according to the present invention1H-1Important correlations between H COSY and HMBC.
FIG. 7 shows Germicidin P (1) in CH obtained from strain 18A01 according to the invention3Experimental ECD spectra in OH, calculated ECD spectra.
FIG. 8 is a HRESIM plot of Germicidin Q (2) obtained using strain 18A01 according to the invention.
FIG. 9 shows Germicidin Q (2) in CH obtained from strain 18A01 according to the invention3Experimental ECD spectra in OH, calculated ECD spectrogram.
FIG. 10 provides an HRESIMS spectrum of Germicidin R (3) obtained using strain 18A01 in accordance with the invention.
FIG. 11 provides an HRESIMS spectrum of Germicidin S (4) obtained using strain 18A01 in accordance with the invention.
Detailed Description
For a better understanding of the present invention, reference will now be made to the following examples, which are included to illustrate, but are not intended to limit the scope of the present invention.
Example 1 identification and characterization of Streptomyces spongiensis 18A01
The strain 18A01 is separated from sponge collected from sea area of Yongxing island of Xisha Genjima, and is preserved in China general microbiological culture collection management center in 12 months and 10 days in 2020 with the preservation number: CGMCC 21351. The strain grew well on TSB, Gao's No. 1, ISP3 medium, forming a round off-white colony without pigment production (FIG. 1).
Streptomyces 18A01 was inoculated into TSB liquid medium containing 2.5% artificial sea salt, cultured at 28 ℃ for 36 hours, and total DNA was extracted according to the conventional method. Then, genome determination and assembly are carried out by adopting a second-generation high-throughput sequencing technology Illumina HiSeq2500 to obtain a genome sketch of the strain 18A01, the genome size is about 8.76Mbp, the GC content is 72.17%, and 54 Scaffol are obtained by assemblyds, N50 size 385,800 bp. BLAST analysis of 16S rRNA gene revealed that it is different from the standard strain S.coelescens DSM 40421 of four different speciesT、S.anthocyanicus NBRC 14892T、 S.violaceoruber NBRC 12826TTricolor NBRC 15461TSequence homology is up to 100%. Phylogenetic trees further constructed based on the 16S rDNA sequences showed that these strains clustered in the same branch (fig. 2), showing that they have the closest genetic evolutionary relationship. Calculation of strains 18A01 and NBRC 15461T(genome size about 8.83Mbp, GC content 72.48%) and results show that Kostas lab calculated ANI between them was 83.66% (int.J.Syst.Evol.Microbiol.,2007,57(Pt1):81-91) and Ezbiocool calculated ANI between them was 82.64% (Antonie van Leeuwenhoek, 2017, 110: 1281-1286). Strain 18a01 was suggested to be a different species from s.tricolor, as indicated by the identity of strain ANI greater than 95%. The summary information shows that 18A01 is taxonomically certain.
Wherein the 16S rRNA gene sequence 16S rDNA of the streptomycete 18A01 is as follows:
>18A01 16S rDNA
ACGATGAACCACTTACGGTGGGGATTAGTGGCGAACGGGTGAGT AACACGTGGGCAATCTGCCCTTCACTCTGGGACAAGCCCTGGAAACG GGGTCTAATACCGGATACTGACCCTCGCAGGCATCTGCGAGGTTCGAA AGCTCCGGCGGTGAAGGATGAGCCCGCGGCCTATCAGCTTGTTGGTG AGGTAATGGCTCACCAAGGCGACGACGGGTAGCCGGCCTGAGAGGG CGACCGGCCACACTGGGACTGAGACACGGCCCAGACTCCTACGGGA GGCAGCAGTGGGGAATATTGCACAATGGGCGAAAGCCTGATGCAGCG ACGCCGCGTGAGGGATGACGGCCTTCGGGTTGTAAACCTCTTTCAGC AGGGAAGAAGCGAAAGTGACGGTACCTGCAGAAGAAGCGCCGGCTA ACTACGTGCCAGCAGCCGCGGTAATACGTAGGGCGCAAGCGTTGTCC GGAATTATTGGGCGTAAAGAGCTCGTAGGCGGCTTGTCACGTCGGTTG TGAAAGCCCGGGGCTTAACCCCGGGTCTGCAGTCGATACGGGCAGGC TAGAGTTCGGTAGGGGAGATCGGAATTCCTGGTGTAGCGGTGAAATG CGCAGATATCAGGAGGAACACCGGTGGCGAAGGCGGATCTCTGGGCC GATACTGACGCTGAGGAGCGAAAGCGTGGGGAGCGAACAGGATTAKA TACCCTGGTAGTCCACGCCGTAAACGGTGGGCACTAGGTGTGGGCAA CATTCCACGTTGTCCGTGCCGCAGCTAACGCATTAAGTGCCCCGCCTG GGGAGTACGGCCGCAAGGCTAAAACTCAAAGGAATTGACGGGGGCC CGCACAAGCGGCGGAGCATGTGGCTTAATTCGACGCAACGCGAAGAA CCTTACCAAGGCTTGACATACACCGGAAAGCATCAGAGATGGTGCCC CCCTTGTGGTCGGTGTACAGGTGGTGCATGGCTGTCGTCAGCTCGTGT CGTGAGATGTTGGGTTAAGTCCCGCAACGAGCGCAACCCTTGTCCCG TGTTGCCAGCAAGCCCTTCGGGGTGTTGGGGACTCACGGGAGACCGC CGGGGTCAACTCGGAGGAAGGTGGGGACGACGTCAAGTCATCATGCC CCTTATGTCTTGGGCTGCACACGTGCTACAATGGCCGGTACAATGAGC TGCGATACCGCAAGGTGGAGCGAATCTCAAAAAGCCGGTCTCAGTTC GGATTGGGGTCTGCAACTCGACCCCATGAAGTCGGAGTCGCTAGTAAT CGCAGATCAGCATTGCTGCGGTGAATACGTTCCCGGGCCTTGTACACA CCGCCCGTCACGTCACGAAAGTCGGTAACACCCGAAGCCGGTGGCCC AACCCCTTGTGGGAGGGAGCTGTCGAAGGTGGGACTGGCGATTGGGA CGAAGTCGTAACAAGGTAGCCGTACCGGAAGGTGCGGCTGGATCACC TCCTTT
then 18A01 was found to harbor more than 30 secondary metabolite gene clusters using anti SMASH analysis and manual correction, with the potential to produce Desferrioxamine B, Geosmin, Isorenieratene, Hopen, Albaflavone, Ectoine, Germicidin, SapB (lanthione), Spore fragment, Calcium-dependent inhibitory (CDA), Coelimycin P1, undercyclic prediiosin, Coelicidin, Melanin, Coelictin and Actinorhodorin backbone compounds, and the potential to synthesize various novel PKS and NRPS classes (Table 1). It was demonstrated that 18A01 is an excellent strain for producing a variety of bioactive lead compounds.
TABLE 1 potential of Strain 18A01 for the Synthesis of diverse Secondary metabolites
Figure RE-GDA0002979895260000061
Example 2 production of various types of bioactive substances by Streptomyces marinus 18A01 and methods of preparation
Further using OSMAC strategy to ferment and improve the chemical diversity of the secondary metabolites produced by 18A01, and through activity screening and HPLC-DAD-MS analysis, the strain can produce a series of secondary metabolites of alpha-pyrone and other types when ISP3 (the components comprise 20 g of oat flour, 1mL of trace elements, 1L of tap water and pH 7.2) is used as a fermentation medium (figure 3); the method specifically comprises the following steps:
inoculating marine streptomyces 18A01 on ISP3 solid culture medium, culturing for 48h, activating, and inoculating on artificial sea saltCulturing the ISP3 liquid culture medium at 28 ℃ for 72h to obtain a seed solution; and then, mixing the fermented seed liquid according to the volume ratio of 1: 18 were inoculated into the same fermentation medium and cultured with shaking at 28 ℃ for 7 days. Centrifuging the fermentation liquor to remove thallus to obtain fermentation supernatant, and adsorbing active ingredients in the fermentation liquor by using macroporous adsorption resin XAD16 solid phase, wherein the volume ratio of the resin to the fermentation liquor is 1: 25; and after the adsorption is finished for 2h, washing the resin with distilled water, desorbing with methanol, and recovering the methanol solvent to obtain a crude extract A. Separating the crude extract A by adopting a flash silica gel column chromatography, and performing separation according to a dichloromethane ratio: gradient eluting with methanol (v/v) ratio of 100:0-0:100 to obtain 4 fractions (F)A-FD). Fraction FARemoving impurities by gel LH20, eluting with 40-55% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain germicidin A (6.0mg, t)R:19.86min),germicidin B(2.7mg,tR:17.95 min),isogermicidin B(0.8mg,tR:18.20min),germicidin C(1.6mg,tR:18.22 min),germicidin D(4.0mg,tR:15.97min),germicidin H(3.6mg,tR16.33 min), ferulic acid (2.5mg, t)R14.40 min). Fraction FBEluting with 35% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain compound germicidin P (4.0 mg, t)R:13.21min),germicidin Q(4.0mg,tR:13.69min),germicidin R(5.2mg, tR:14.04min),germicidin S(4.3mg,tR:14.35min),germicidin L(9.7mg,tR: 12.58min),germicidin M(3.6mg,tR:12.85min),(1S,2S)-germicidin N(22.4 mg,tR14.90 min). Fraction FCRemoving impurities by gel LH20, eluting with 25% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain compound alpha-ribavirin (5.9 mg, t)R10.53 min). Fraction FDRemoving impurities by gel LH20, eluting with 30% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain compound N-acetyltryptophan (17.3mg, t)R14.29 min). The structure of the compound is shown in figure 4.
The structure of the compound obtained above was analyzed, and specifically, Compound 1 was a yellow oily substanceHRESIMS data show an M/z 199.0965 [ M + H ]]+Ion peak (FIG. 5), corresponding to molecular formula C10H14O4The unsaturation degree is 4. The ultraviolet absorption at 290nm in the ultraviolet spectrum is the characteristic absorption of alpha-pyrone.1H NMR(600MHz,DMSO-d6) Showing the presence of an olefinic hydrogen proton deltaH6.01(s, H-5), two methylene protons, δH2.42(m, H-7) and 2.25(d, J ═ 14.7,7.3Hz, H-10), one methine proton δH3.89(m, H-8), one doublet methyl deltaH1.08(d, J ═ 6.2Hz, H-9) and one triplet methyl deltaH 0.94(t,J=14.7,7.3Hz,H-11)。13C NMR (600MHz,DMSO-d6) Shows 5 sp2Hybrid carbon and 5 aliphatic carbons. H-10 and C-4 (. delta.) in HMBC spectraC164.7), 4-OH and C-3 (. delta.))C102.8), H-11 and C-3 (. delta.))C102.8) and H-5 and C-3 (. delta.))C102.8) concluded the presence of a 4-hydroxy-3-ethyl-alpha-pyrone backbone (FIG. 6). Linkage of H-7, H-8 and H-9 in COSY spectra and H-7 with C-9 (. delta.) in HMBC spectraC23.5) concluded that a 2-hydroxy-propyl group is present (FIG. 6). H-7 and C-5 (. delta.) in HMBC spectraC100.9) and C-6 (. delta.))C160.8) that the 2-hydroxy-propyl group is attached to C-6 (. delta.) of the 4-hydroxy-3-ethyl-. alpha. -pyrone skeletonC160.8). Thus, the planar structure of compound 1 is 4-hydroxy-3-ethyl-6- (2-hydroxy-propyl) - α -pyrone. The absolute configuration of compound 1 obtained by calculation of ECD was 8R (fig. 7). Compound 1 was therefore designated as a novel compound germicidin P (figure 4).
Compound 2 was a yellow oil and HRESIMS data showed an M/z 199.09634 [ M + H ]]+Ion peak (FIG. 8) with molecular formula C10H14O4The unsaturation degree is 4. The ultraviolet absorption at 290nm in the ultraviolet spectrum is the characteristic absorption of alpha-pyrones. The 1D NMR spectrum showed that Compound 2 was structurally similar to Compound 1 except for the substituent at the C-6 position. Linkage of H-7, H-8 and H-9 in COSY spectra and H-9 to C-8 (. delta.) in HMBC spectraC63.5) concluded the presence of a 2-hydroxy-1-methyl-ethyl group (FIG. 6). H-7 and C-5 (. delta.) in HMBC spectraC99.4) and C-6 (. delta.))C164.8) can be connectedKnowing that the 2-hydroxy-1-methyl-ethyl group is attached to the C-6 (. delta.) -3-ethyl-. alpha. -pyrone skeletonC160.8). Thus, the planar structure of compound 2 is 4-hydroxy-3-ethyl-6- (2-hydroxy-1-methyl-ethyl) - α -pyrone. The absolute configuration of compound 2 obtained by the calculation of ECD was 7S (fig. 9). Compound 2 was therefore named germicidin Q as a novel compound (figure 4).
Compound 3 was a yellow oil, with HRESIMS data showing an M/z 199.09636 [ M + H ]]+Ion peak (FIG. 10) with molecular formula C10H14O4The unsaturation degree is 4. The ultraviolet absorption at 290nm in the ultraviolet spectrum is the characteristic absorption of alpha-pyrone. The 1D NMR spectrum shows that the compound 3 has a structure similar to that of the compound germicidin B7, and is composed of C-7 (delta)C69.5) the difference is that the hydrogen in position 7 is replaced by a hydroxyl group. The planar structure of compound 3 was inferred to be 4-hydroxy-3-ethyl-6- (1-hydroxy-1-methyl-ethyl) - α -pyrone by the linkage of COSY spectrum and HMBC spectrum (fig. 6). Compound 3 was therefore named germicidin R as a novel compound (figure 4).
Compound 4 was a yellow oil and HRESIMS data showed an M/z 213.11220 [ M + H ]]+Ion peak (FIG. 11) with molecular formula C11H16O4The unsaturation degree is 4. The ultraviolet absorption at 290nm in the ultraviolet spectrum is the characteristic absorption of alpha-pyrone. The 1D NMR spectrum showed that Compound 4 was structurally similar to Compound 1, consisting of C-9 and C-10 (. delta.) (delta.)C29.4) the difference is that the hydrogen in position 8 is replaced by a methyl group. The planar structure of compound 4 was inferred to be 4-hydroxy-3-ethyl-6- (2-hydroxy-2-methyl-propyl) - α -pyrone by the linkage of COSY spectrum and HMBC spectrum (fig. 6). Compound 4 was therefore designated as a novel compound germicidin S (figure 4). The 1D-NMR data of the above 4 novel compounds are shown in tables 2 and 3.
TABLE 2 preparation of Compounds 1 to 41H NMR data (600MHz, DMSO-d)6,δin ppm,J in Hz)
Figure RE-GDA0002979895260000081
TABLE 3 Compounds1-4 of13C NMR data (150MHz, DMSO-d)6)
Figure RE-GDA0002979895260000082
Figure RE-GDA0002979895260000091
Other known compounds, germicidin A (5), germicidin B (6), isogermicidin B (7), germicidin C (8), germicidin D (9), germicidin H (10), germicidin L (11), germicidin M (12), (1S,2S) -germicidin N (13), ferulic acid (14), alpha-ribazole (15) and N-acetyltryptophan (16) (FIG. 4), were determined from literature relevant to comparative reports of data such as 1D-NMR spectra and optical rotation.
Example 3 Streptomyces marinus 18A01 and alpha-pyrone Compounds have HK2 inhibitory Activity
HK2 is the rate-limiting enzyme in the first step of the glycolysis pathway and is expressed at higher levels in cancer cells than in normal cells. HK2 provides a new target for tumor therapy due to its key role in tumorigenesis and metastasis.
The extract of the marine streptomyces 18A01 and the compound prepared by separation are used for measuring the inhibition effect of the extract on HK2 by using an HK2 enzyme activity measuring kit. HK2 catalyzes the phosphorylation of glucose to glucose-6-phosphate, which is oxidized by glucose-6-phosphate dehydrogenase to form NADPH. The production rate of NADPH is related to glucose concentration, NADPH has specific absorption peak at 340nm, absorbance change is detected at 340nm, and IC of inhibitory activity of compound on HK2 is determined50Values, each set of experiments was performed in triplicate. Test results show that the alpha-pyrone compounds germicidins obtained by separation exhibit inhibitory activity to HK2 to different degrees, and specifically comprise the following components:
the compounds germicidins P, Q, R and (1S,2S) -germicidin N have stronger inhibitory activity on HK2, and particularly the compounds germicidin Q and (1S,2S) -germicidin N inhibit IC of HK250Values of 5.13 and 5.30. mu.M, respectively, and positivityIC of control drug benserazide50The values are approximately the same (IC)505.52 μ M), both of them are promising candidates for HK2 inhibitors.
TABLE 4.18A 01 extracts and their alpha-pyrones prepared inhibit the activity of HK2
Compd. IC50 Compd. IC50 a
germicidin P(1) 8.20±0.21 germicidin H(10) 39.51±1.34
germicidin Q(2) 5.13±0.53 germicidin L(11) 10.55±0.84
germicidin R(3) 7.84±0.78 germicidin M(12) 11.03±0.67
germicidin S(4) 10.27±0.69 (1S,2S)-germicidin N(13) 5.30±0.64
germicidin A(5) 19.33±0.74 benserazideb 5.52±0.17
germicidin B(6) 25.16±1.10 18A01 extractc 0.784μg/mL
germicidin D(9) 32.38±0.44
a IC50 values represent the means±SD of three parallel measurements,μM.
b positive control.c μg/mL.
Example 4 anti-tumor cell Activity of alpha-pyrone germicidins Compounds
In-vitro cell activity proliferation inhibition experiment, the effect of the compound on the inhibition rate of each cell is examined by adopting a CCK-8 detection method.
Digesting each cell in logarithmic growth phase with pancreatin, collecting, mixing, counting, and adjusting density to 1 × 105Cells per mL were seeded at 100. mu.l/well in 96-well plates and cultured overnight (5% CO) at 37 ℃2100% humidity); the stock culture was then discarded and given serum-free blank medium containing different final concentrations of germicidins (0-40. mu.M) for 72h (3 replicates per concentration). Then 10. mu.l of CCK-8 solution 5mg/mL are addedWells, after 2h incubation. Measuring its light absorbance at 570nm with microplate reader, and calculating half inhibition concentration IC of each cell after administration50. Experimental results show that germicidin R has activity (IC) against colon cancer HCT116 cells50=15.35±1.56μM)。
Sequence listing
<110> Shenyang application ecological research institute of Chinese academy of sciences
<120> preparation and application of streptomyces spongiensis and substance for inhibiting hexokinase II activity
<160> 1
<170> SIPOSequenceListing 1.0
<210> 1
<211> 1463
<212> DNA
<213> Artificial Sequence (Artificial Sequence)
<400> 1
acgatgaacc acttacggtg gggattagtg gcgaacgggt gagtaacacg tgggcaatct 60
gcccttcact ctgggacaag ccctggaaac ggggtctaat accggatact gaccctcgca 120
ggcatctgcg aggttcgaaa gctccggcgg tgaaggatga gcccgcggcc tatcagcttg 180
ttggtgaggt aatggctcac caaggcgacg acgggtagcc ggcctgagag ggcgaccggc 240
cacactggga ctgagacacg gcccagactc ctacgggagg cagcagtggg gaatattgca 300
caatgggcga aagcctgatg cagcgacgcc gcgtgaggga tgacggcctt cgggttgtaa 360
acctctttca gcagggaaga agcgaaagtg acggtacctg cagaagaagc gccggctaac 420
tacgtgccag cagccgcggt aatacgtagg gcgcaagcgt tgtccggaat tattgggcgt 480
aaagagctcg taggcggctt gtcacgtcgg ttgtgaaagc ccggggctta accccgggtc 540
tgcagtcgat acgggcaggc tagagttcgg taggggagat cggaattcct ggtgtagcgg 600
tgaaatgcgc agatatcagg aggaacaccg gtggcgaagg cggatctctg ggccgatact 660
gacgctgagg agcgaaagcg tggggagcga acaggattak ataccctggt agtccacgcc 720
gtaaacggtg ggcactaggt gtgggcaaca ttccacgttg tccgtgccgc agctaacgca 780
ttaagtgccc cgcctgggga gtacggccgc aaggctaaaa ctcaaaggaa ttgacggggg 840
cccgcacaag cggcggagca tgtggcttaa ttcgacgcaa cgcgaagaac cttaccaagg 900
cttgacatac accggaaagc atcagagatg gtgcccccct tgtggtcggt gtacaggtgg 960
tgcatggctg tcgtcagctc gtgtcgtgag atgttgggtt aagtcccgca acgagcgcaa 1020
cccttgtccc gtgttgccag caagcccttc ggggtgttgg ggactcacgg gagaccgccg 1080
gggtcaactc ggaggaaggt ggggacgacg tcaagtcatc atgcccctta tgtcttgggc 1140
tgcacacgtg ctacaatggc cggtacaatg agctgcgata ccgcaaggtg gagcgaatct 1200
caaaaagccg gtctcagttc ggattggggt ctgcaactcg accccatgaa gtcggagtcg 1260
ctagtaatcg cagatcagca ttgctgcggt gaatacgttc ccgggccttg tacacaccgc 1320
ccgtcacgtc acgaaagtcg gtaacacccg aagccggtgg cccaacccct tgtgggaggg 1380
agctgtcgaa ggtgggactg gcgattggga cgaagtcgta acaaggtagc cgtaccggaa 1440
ggtgcggctg gatcacctcc ttt 1463

Claims (6)

1. A Streptomyces spongiensis is characterized in that: the marine streptomyces isStreptomycessp.18A01, deposited in China general microbiological culture Collection center at 12 months and 10 days in 2020, with the deposit number: CGMCC 21351.
2. The Streptomyces spongiensis according to claim 1, wherein: the marine streptomyces contains an 8.76Mbp genome; the genome contains clusters of genes that synthetically produce Desferrioxamine B, Geosmin, Isorenieratene, Hopen, Albaflavone, Germidin, SapB (lanthionide), Spore fragment, Calcium-dependent antibody (CDA), Coelimycin P1, Undeclyprodigoside, Coelicidin, Melanin, Coelictin and Actinorhoddin backbone compounds.
3. Use of streptomyces spongiensis according to claim 1, characterized in that: the metabolic product of the marine streptomyces 18A01 is used as a hexokinase II inhibitor or an anti-tumor candidate drug.
4. Use of streptomyces spongiensis according to claim 1, characterized in that: the metabolic product of the marine streptomyces 18A01 is used for preparing alpha-pyrone germicidins, ferulic acid,α-ribazole andN-any compound of acetyltryptophan.
5. Use of streptomyces spongiensis according to claim 3 or 4, characterized in that: the preparation of the marine streptomyces 18A01 metabolite comprises the following steps:
1) inoculating the marine streptomyces 18A01 of claim 1 on an ISP3 solid culture medium for 2-5 days of activation, then inoculating on an ISP3 liquid culture medium containing artificial sea salt, and culturing at 28 ℃ for 2-4 days as a seed solution; inoculating the fermented seed liquid 2-10% by volume to ISP3 liquid culture medium containing artificial sea salt, and performing shaking culture at 28 deg.C for 5-10 d;
2) centrifuging the fermentation liquor to remove thallus to obtain fermentation supernatant, and adsorbing active ingredients in the fermentation liquor by using macroporous adsorption resin XAD16 solid phase, wherein the volume percentage of the resin to the fermentation liquor is 3% -6%; after adsorbing for 2h-4h, washing the resin with distilled water, desorbing with methanol, and recovering the methanol solvent to obtain crude extract A, namely the metabolic product of Streptomyces marinus 18A 01.
6. Use of Streptomyces spongiensis according to claim 5, characterized in that: further purifying and preparing the crude extract A to obtain germicidins, ferulic acid,α-ribazole andN-acetyltryptophan: separating the crude extract A by adopting a flash silica gel column chromatography, and performing separation according to a dichloromethane ratio: methanol (c)v/v) Gradient eluting at a ratio of 100:0-0:100 to obtain 4 fractions (F)A-FD) (ii) a Fraction FBAdopting semi-preparative reverse phase HPLC, eluting with 35% methanol water solution at the flow rate of 2.5mL/min to obtain compounds germicidin P (1), germicidin Q (2), germicidin R (3), germicidin S (4), germicidin L (11), germicidin M (12), (1)S, 2S) -germicidin N (13); fraction FARemoving impurities by gel LH20, adopting semi-preparative reverse phase HPLC, eluting by 40-55% methanol water solution with the flow rate of 2.5mL/min, and obtaining compounds germicidin A (5), germicidin B (6), isogermicidin B (7), germicidin C (8), germicidin D (9), germicidin H (10) and ferulic acid (14); fraction FCRemoving impurities by gel LH20, performing semi-preparative reverse phase HPLC, eluting with 25% methanol water solution at flow rate of 2.5mL/min to obtain compoundα-a ribazole (15); fraction FDRemoving impurities by gel LH20, eluting with 30% methanol water solution by semi-preparative reverse phase HPLC at flow rate of 2.5mL/min to obtain compoundN-acetyltryptophan (16)。
CN202011604925.9A 2020-12-30 2020-12-30 Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II Active CN112725227B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011604925.9A CN112725227B (en) 2020-12-30 2020-12-30 Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011604925.9A CN112725227B (en) 2020-12-30 2020-12-30 Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II

Publications (2)

Publication Number Publication Date
CN112725227A CN112725227A (en) 2021-04-30
CN112725227B true CN112725227B (en) 2022-07-05

Family

ID=75610790

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011604925.9A Active CN112725227B (en) 2020-12-30 2020-12-30 Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II

Country Status (1)

Country Link
CN (1) CN112725227B (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103436458B (en) * 2013-04-24 2015-06-24 中国科学院沈阳应用生态研究所 Streptomyces, spiroteteronate compound produced by streptomyces, and preparation method and application of spiroteteronate compound
CN110563740B (en) * 2019-08-05 2020-10-02 浙江工业大学 Alpha-pyrone compound, preparation method, strain and application

Also Published As

Publication number Publication date
CN112725227A (en) 2021-04-30

Similar Documents

Publication Publication Date Title
CN107298671B (en) Selenolonic acid H from penicillium oxalicum and application thereof in preparing medicine for resisting human colon cancer
Supong et al. Antimalarial and antitubercular C-glycosylated benz [α] anthraquinones from the marine-derived Streptomyces sp. BCC45596
CN107298672B (en) Application of seclenic acid I derived from penicillium oxalicum in preparation of anti-human colon cancer drugs
US7745198B2 (en) Method of producing macrolide compound
JPS6023679B2 (en) Rhodomycin group antibiotics and their production method
AU634247B2 (en) Antitumor substance be-13793c
CN107485607B (en) Application of seclenic acid H derived from penicillium oxalicum in preparation of human esophageal cancer resistant medicine
CN107298670B (en) Application of medicine derived from penicillium oxalicum seclenum ketonic acid H in preparation of anti-human oral epidermoid carcinoma medicines
CN112725227B (en) Sponge-derived streptomyces and preparation and application of active substance for inhibiting hexokinase II
CN114213428B (en) Indole alkaloid compound and preparation method and application thereof
US20130230890A1 (en) Pseudonocardia SP. And Method For Preparing Deoxynyboquinone By Utilizing Same
JP5826406B2 (en) Streptomyces, antitumor compound Spiro-Indymycin AD, production method and use thereof, and antitumor agent and drug containing spiroindimycin
CN112500348B (en) Geldanamycin derivatives, preparation method thereof and application thereof in preparing antitumor drugs
CN109134416B (en) Application of seclenic acid H derived from penicillium oxalicum in preparation of human cervical cancer drugs
CN102351859B (en) Antibiotic Pseudonocardian A and Pseudonocardian B, its preparation method thereof and its application in preparation of antibiotics and antitumor drug
JPH1017593A (en) New peptide, its production and inhibitor of cell cycle comprising the same
Bunbamrung et al. Anthrone derivatives from the terrestrial actinomycete, Actinomadura sp. BCC47066
CN111944702B (en) Preparation method and application of aromatic butenolide
CN113861208A (en) Cytochalasin compound and preparation method and application thereof
US8952140B2 (en) Compound amycolose derivative, method for producing the same, and use of the same
US7439225B2 (en) Substances K01-B0171 and process for producing the same
EP0213320B1 (en) Physiologically active formamides
CN114891017B (en) Maleic anhydride alicyclic compound and preparation method and application thereof
JP5641716B2 (en) Novel compounds produced by microorganisms belonging to the genus Streptomyces, microorganisms producing the compounds, and pharmaceutical preparations containing the compounds as active ingredients
CN114605430B (en) Macrocyclic dilactone compound, and preparation method 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